WO2020165834A1 - Dérivés de 3-(1-oxoisoindoline-2-yl)pipéridine-2,6-dione substitués et leurs utilisations - Google Patents

Dérivés de 3-(1-oxoisoindoline-2-yl)pipéridine-2,6-dione substitués et leurs utilisations Download PDF

Info

Publication number
WO2020165834A1
WO2020165834A1 PCT/IB2020/051206 IB2020051206W WO2020165834A1 WO 2020165834 A1 WO2020165834 A1 WO 2020165834A1 IB 2020051206 W IB2020051206 W IB 2020051206W WO 2020165834 A1 WO2020165834 A1 WO 2020165834A1
Authority
WO
WIPO (PCT)
Prior art keywords
ikzf2
pharmaceutically acceptable
formula
prodrug
solvate
Prior art date
Application number
PCT/IB2020/051206
Other languages
English (en)
Inventor
Rohan Eric John Beckwith
Simone BONAZZI
Artiom CERNIJENKO
Philip Lam
Noel Marie-France THOMSEN
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to EA202192029A priority Critical patent/EA202192029A1/ru
Priority to CA3123519A priority patent/CA3123519A1/fr
Priority to US17/430,478 priority patent/US20220144798A1/en
Priority to KR1020217028320A priority patent/KR20210129672A/ko
Priority to MX2021009764A priority patent/MX2021009764A/es
Priority to CN202080009260.9A priority patent/CN113329792A/zh
Priority to JP2021547485A priority patent/JP2022520448A/ja
Priority to AU2020222346A priority patent/AU2020222346B2/en
Priority to EP20708627.3A priority patent/EP3924055B1/fr
Publication of WO2020165834A1 publication Critical patent/WO2020165834A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present disclosure relates to substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione compounds and compositions and their use for the treatment of IKAROS Family Zinc Finger 2 (IKZF2)- 10 dependent diseases or disorders or where reduction of IKZF2 or IKZF4 protein levels can ameliorate a disease or disorder.
  • IKAROS Family Zinc Finger 2 IKZF2
  • IKZF4 protein levels can ameliorate a disease or disorder.
  • IKZF2 IKAROS Family Zinc Finger 2
  • IKZF2 contains four zinc finger domains near the 15 N-terminus, which are involved in DNA binding, and two zinc finger domains at the C-terminus, which are involved in protein dimerization.
  • IKZF2 is about 50% identical with Ikaros family members, Ikaros (IKZF1), Aiolos (IKZF3), and Eos (IKZF4) with highest homology in the zinc finger regions (80%+ identity).
  • Ikaros family transcription factors bind to the same DNA consensus site and can heterodimerize with each other when co-expressed in cells.
  • the fifth Ikaros family protein, Pegasus 20 (IKZF5), is only 25% identical to IKZF2, binds a different DNA site than other Ikaros family members and does not readily heterodimerize with the other Ikaros family proteins.
  • IKZF2, IKZF1 and IKZF3 are expressed mainly in hematopoietic cells while IKZF4 and IKZF5 are expressed in a wide variety of tissues.
  • IKZF2 is believed to have an important role in the function and stability of regulatory T cells (Tregs). IKZF2 is highly expressed at the mRNA and protein level by regulatory T-cell populations. Knockdown of IKZF2 by siRNA has been shown to result in downregulation of FoxP3 and to impair the ability of isolated human CD4+ CD25+ Tregs to block T-cell activation in vitro. Moreover, overexpression of IKZF2 in isolated murine Tregs has been shown to increase expression of Treg related markers such as 30 CD103 and GITR and the IKZF2 overexpressing cells showed increased suppression of responder T-cells.
  • Treg related markers such as 30 CD103 and GITR
  • IKZF2 has also been found to bind the promoter of FoxP3, the defining transcription factor of the regulatory T-cell lineage, and to affect FoxP3 expression.
  • IKZF2 knockout mutant mice develop autoimmune disease by 6-8 months of age, with increased numbers of activated CD4 and CD8 T cells, follicular helper T cells and germinal center B cells. This observed effect is believed to be cell intrinsic, as Rag2-/- mice given bone marrow from IKZF2 knockout mice, but not bone marrow from IKZF2+/+ develop autoimmune disease.
  • IKZF2 affects regulatory T-cell function
  • mice in which IKZF2 was deleted only in FoxP3 expressing cells FoxP3-YFP-Cre Heliosfl/fl.
  • the results showed that the mice also develop autoimmune disease with similar features as observed in the whole animal IKZF2 knockout.
  • pathway analysis 5 of a CHIP-SEQ experiment has also suggested that IKZF2 is affecting expression of genes in the STAT5/IL-2Ra pathway in regulatory T-cells.
  • Ikaros isoforms which lack the DNA binding regions have been shown to be associated with multiple human haematological malignancies. Recently, mutations in the IKZF2 gene, which lead to abnormal splicing variants, have been identified in adult T-cell leukemias and low 15 hypodiploid acute lymphoblastic leukemia. It has been proposed that these isoforms, which are capable of dimerization, have a dominant negative effect on Ikaros family transcription factors which primes the development of lymphomas. IKZF2 knockout mutants that survive into adulthood do not develop lymphomas, supporting this hypothesis (Asanuma, S., et al., (2013), Cancer Sci.
  • An IKZF2-specific degrader has the potential to focus the enhanced immune response to areas within or near tumors providing a potentially more tolerable and less toxic therapeutic agent for the treatment of cancer.
  • the compounds of the disclosure have use as therapeutic agents, particularly for cancers and related 30 diseases.
  • the compounds of the disclosure have IKZF2 degrader activity, preferably having such activity at or below the 50 ⁇ M level, and more preferably having such activity at or below the 10 ⁇ M level.
  • the compounds of the disclosure have degrader activity for IKZF2 that is selective over one or more of IKZF1, IKZF3, IKZF4, and/or IKZF5.
  • the compounds of the disclosure have degrader activity for both IKZF2 and IKZF4.
  • the compounds of the disclosure have 35 usefulness in treating cancer and other diseases for which such degrader activity would be beneficial for the patient.
  • the present disclosure provides novel IKZF2 degraders useful for the treatment of cancer and other diseases.
  • a first aspect of the present disclosure relates to compounds of Formula (I)
  • X 1 is CR 3 ;
  • X 2 is N and X 3 is CR 14 ; or X 2 is CR 13 and X 3 is N; or X 2 is CR 15 and X 3 is CR 14 ; or X 2 is CR 13 and X 3 is CR 16 ;
  • each R 1 is independently D, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )hydroxyalkyl, CN, or halogen, or two R1 together with the carbon atoms to which they are attached form (C3-C7)cycloalkyl or a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, or two R 1 , when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S; 15 R 2 is (C 1 -C 6 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )
  • R 1 and R 2 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6- membered heterocycloalkyl ring;
  • R 3 is H or D, or R 3 is absent when s a double bond
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, -OH, -NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, 25 (C 3 -C 8 )cycloalkyl, and 4- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms
  • aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one or more R 7 ;
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 3 -C 7 )cycloalkyl, 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 6 -C 10 )aryl, and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, or
  • R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl 5 ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 , or
  • R6 and R6' are each independently H, (C 1 -C 6 )alkyl, or (C6-C10)aryl;
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -(CH 2 ) 0-3 C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , - NR 8 C(O)OR 9 , -S(O) p NR 8 R 9 , -S(O) p R 12 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, -O(CH 2 ) 0-3 (C 6 -C 10 )aryl, adam
  • R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 , or
  • R 7 together with the atoms to which they are attached form a (C 5 -C 7 ) cycloalkyl ring or a 5- to 7- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 ;
  • R 8 and R 9 are each independently H or (C 1 -C 6 )alkyl
  • each R 10 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl,
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, (C 6 -C 10 )aryl, and 5- to 7-membered
  • heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heterocycloalkyl are optionally substituted with one or more substituents each independently selected 35 from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN;
  • R 12 is (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 6 -C 10 )aryl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S;
  • R13 is H, halogen, -OH, or -NH2;
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, 5 halogen, -OH, -NH 2 , -NO 2 , or CN;
  • R 15 is halogen, -OH, or -NH 2 ;
  • R 16 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN;
  • R x is H or D
  • 10 p is 0, 1, or 2;
  • n 0, 1, or 2;
  • n1 is 1 or 2, wherein n + n1 £ 3;
  • q 0, 1, 2, 3, or 4;
  • the hydrogens in the compound of Formula (I) are present in their normal isotopic abundances.
  • the hydrogens are isotopically enriched in deuterium (D), and in a particularly preferred aspect of the invention the hydrogen at position R x is enriched in D, as discussed in more detail concerning isotopes and isotopic enrichment below.
  • compositions comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is useful in the treatment of IKZF2-dependent diseases or disorders.
  • the pharmaceutical composition may further comprise at least one additional pharmaceutical 25 agent.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable carrier or excipient for use in the treatment of an IKZF2-dependent disease or disorder by reducing IKZF2 protein 30 levels wherein reduction of IKZF2 protein levels treats the IKZF2-dependent disease or disorder.
  • the pharmaceutical composition is useful in the treatment of IKZF2-dependent diseases or disorders.
  • the pharmaceutical composition may further comprise at least one additional pharmaceutical agent.
  • compositions comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, 35 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition is useful in the treatment of diseases or disorders affected by the reduction of IKZF2 protein levels.
  • the pharmaceutical composition may further comprise at least one additional pharmaceutical agent.
  • the present disclosure relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, 5 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable carrier or excipient for use in the treatment of a disease or disorder affected by the reduction of IKZF2 protein levels wherein reduction of IKZF2 protein levels treats the disease or disorder.
  • the pharmaceutical composition may further comprise at least one additional pharmaceutical agent.
  • Another aspect of the present disclosure relates to a method of degrading IKZF2 comprising 10 administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a method of treating a disease or disorder that is affected by the modulation of IKZF2 protein levels comprising administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, 15 or tautomer thereof.
  • Another aspect of the present disclosure relates to a method of modulating IKZF2 protein levels comprising administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a method of reducing the proliferation of a cell 20 the method comprising, contacting the cell with a compound of Formula (I), or a pharmaceutically
  • Another aspect of the present disclosure relates to a method of treating cancer comprising administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable 25 salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the cancer is
  • the cancer is a cancer for which the immune response is deficient or an immunogenic cancer.
  • the present disclosure relates to a method for reducing IKZF2 protein levels in a subject comprising the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in 35 the treatment of a disease or disorder that is affected by the reduction of IKZF2 protein levels.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating a disease or disorder that is affected by the reduction of IKZF2 protein levels.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in 5 the manufacture of a medicament for treating a disease or disorder associated with the reduction of IKZF2 protein levels.
  • the disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and
  • GIST gastrointestinal stromal tumor
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of a disease or disorder associated with the reduction of IKZF2 protein levels.
  • the disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal 15 cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and gastrointestinal stromal tumor (GIST).
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • NPC nasopharyngeal cancer
  • mssCRC microsatellite stable colorectal 15 cancer
  • thymoma carcinoid
  • acute myelogenous leukemia and gastrointestinal
  • the compounds according to the disclosure are formulated into pharmaceutical compositions comprising an effective amount, preferably a pharmaceutically effective amount, of a compound according to the disclosure or salt, hydrate, solvate, prodrug, stereoisomer, or 20 tautomer thereof, and a pharmaceutically acceptable excipient or carrier.
  • the administration of the compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof is performed orally, parentally, subcutaneously, by injection, or by infusion.
  • the present disclosure provides degraders of IKZF2 that are therapeutic agents in the treatment of 25 diseases such as cancer and metastasis, in the treatment of diseases affected by the modulation of IKZF2 protein levels, and in the treatment IKZF2-dependent diseases or disorders.
  • the disease or disorder that can be treated by the compounds of the present disclosure is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, 30 carcinoid, gastrointestinal stromal tumor (GIST), prostate cancer, breast carcinoma, lymphomas, leukaemia, myeloma, bladder carcinoma, colon cancer, cutaneous melanoma, hepatocellular carcinoma, endometrial cancer, ovarian cancer, cervical cancer, lung cancer, renal cancer, glioblastoma multiform, glioma, thyroid cancer, parathyroid tumor, nasopharyngeal cancer, tongue cancer, pancreatic cancer, esophageal cancer, cholangiocarcinoma, gastric cancer, soft tissue sarcomas, rhabdomyosarcoma (RMS), syn
  • the IKZF2-dependent disease or disorder is a cancer for which the immune response is deficient or an immunogenic cancer.
  • the present disclosure provides agents with novel mechanisms of action toward IKZF2 proteins in the treatment of various types of diseases including cancer and metastasis, in the treatment of diseases affected by the modulation of IKZF2 protein levels, and in the treatment IKZF2-dependent diseases or disorders.
  • the present disclosure provides the medical community with a novel pharmacological 5 strategy for the treatment of diseases and disorders associated with IKZF2 proteins.
  • the present disclosure provides agents with novel mechanisms of action toward IKZF2 proteins in the treatment of various types of diseases including cancer and metastasis, in the treatment of diseases affected by the modulation of IKZF2 protein levels, and in the treatment IKZF2-dependent diseases or disorders.
  • the present disclosure provides the medical community with a novel pharmacological 10 strategy for the treatment of diseases and disorders associated with IKZF2 proteins.
  • the present disclosure relates to compounds and compositions that are capable of modulating IKZF2 protein levels.
  • the disclosure features methods of treating, preventing, or ameliorating a disease or disorder in which IKZF2 plays a role by administering to a patient in need thereof a therapeutically effective 15 amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the methods of the present disclosure can be used in the treatment of a variety of IKZF2-dependent diseases and disorders by modulating IKZF2 protein levels. Modulation of IKZF2 protein levels through degradation provides a novel approach to the treatment, prevention, or amelioration of diseases including, but not limited to, cancer and metathesis, and other IKZF2-dependent 20 diseases or disorders.
  • the compounds of the disclosure have use as therapeutic agents, particularly for cancers and related diseases.
  • the compounds of the disclosure have IKZF2 degradation activity, preferably having such activity at or below the 50 ⁇ M level, and more preferably having such activity at or below the 10 ⁇ M level.
  • the compounds of the disclosure have degrader 25 activity for IKZF2 that is selective over one or more of IKZF1, IKZF3, IKZF4, and/or IKZF5.
  • the compounds of the disclosure have degrader activity for both IKZF2 and IKZF4. The compounds of the disclosure have usefulness in treating cancer and other diseases for which such degradation activity would be beneficial for the patient.
  • the present disclosure provides novel IKZF2 degraders useful for the treatment of cancer and other diseases.
  • R 1 , R 2 , R x , X 1 , X 2 , X 3 , n, n1,and q are as defined herein.
  • (C 1 -C 10 )alkyl means an alkyl group or radical having 1 to 10 carbon 20 atoms.
  • the last named group is the radical attachment point, for example,“alkylaryl” means a monovalent radical of the formula alkyl-aryl-, while “arylalkyl” means a monovalent radical of the formula aryl-alkyl-.
  • “alkylaryl” means a monovalent radical of the formula alkyl-aryl-
  • arylalkyl means a monovalent radical of the formula aryl-alkyl-.
  • designating a monovalent radical where a divalent radical is appropriate shall be construed to designate the respective divalent radical and vice versa.
  • an alkyl group that is optionally substituted can be a fully saturated alkyl chain (e.g., a pure hydrocarbon).
  • the same optionally substituted alkyl group can have substituents different from hydrogen. For instance, it can, at any point along the chain be bounded to a halogen atom, a hydroxyl group, or any other substituent described herein.
  • the term“optionally substituted” means that a given chemical moiety has the potential to contain other functional groups, but does not necessarily have any further functional groups.
  • Suitable substituents used in the optional substitution of the described groups include, without limitation, halogen, oxo, -OH, -CN, -COOH, -CH 2 CN, -O-(C 1 -C 6 )alkyl, (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -O-(C 2 -C 6 )alkenyl, -O-(C 2 -C 6 )alkynyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, -OH, - OP(O)(OH) 2 , -OC(O)(C 1 -C 6 )alkyl, -C(O)(C 1 -C 6 )alkyl, -OC(O)O(C 1 -
  • substituted means that the specified group or moiety bears one or more suitable substituents wherein the substituents may connect to the specified group or moiety at one or more positions.
  • an aryl substituted with a cycloalkyl may indicate that the cycloalkyl connects to one atom of 15 the aryl with a bond or by fusing with the aryl and sharing two or more common atoms.
  • aryl means a cyclic, aromatic hydrocarbon group having 1 to 3 aromatic rings, including monocyclic or bicyclic groups such as phenyl, biphenyl, or naphthyl.
  • aromatic rings of the aryl group are optionally joined at 20 a single point (e.g., biphenyl), or fused (e.g., naphthyl).
  • the aryl group is optionally substituted by one or more substituents, e.g., 1 to 5 substituents, at any point of attachment.
  • substituents include, but are not limited to, -H, -halogen, -CN, -O-(C 1 -C 6 )alkyl, (C 1 -C 6 )alkyl, -O-(C 2 -C 6 )alkenyl, -O-(C 2 -C 6 )alkynyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, -OH, -OP(O)(OH) 2 , -OC(O)(C 1 -C 6 )alkyl, -C(O)(C 1 -C 6 )alkyl, - OC(O)O(C 1 -C 6 ) alkyl, NH 2 , NH((C 1 -C 6 )alkyl), N((C 1 -C 6 )alkyl) 2 , -S(O) 2 -(C 1 -C 6 )alky
  • the substituents are themselves optionally substituted.
  • the aryl groups when containing two fused rings, optionally have an unsaturated or partially saturated ring fused with a fully saturated ring.
  • Exemplary ring systems of these aryl groups include, but are not limited to, phenyl, biphenyl, naphthyl, anthracenyl, phenalenyl, phenanthrenyl, indanyl, indenyl, tetrahydronaphthalenyl, tetrahydrobenzoannulenyl, and the like.
  • heteroaryl means a monovalent monocyclic aromatic radical of 5 to 24 ring atoms or a polycyclic aromatic radical, containing one or more ring heteroatoms selected from N, O, or S, the remaining ring atoms being C.
  • Heteroaryl as herein defined also means a bicyclic heteroaromatic group wherein the heteroatom is selected from N, O, or S.
  • the aromatic radical is optionally substituted independently with one or more substituents described herein.
  • Examples include, but 35 are not limited to, furyl, thienyl, pyrrolyl, pyridyl, pyrazolyl, pyrimidinyl, imidazolyl, isoxazolyl, oxazolyl, oxadiazolyl, pyrazinyl, indolyl, thiophen-2-yl, quinolyl, benzopyranyl, isothiazolyl, thiazolyl, thiadiazole, indazole, benzimidazolyl, thieno[3,2-b]thiophene, triazolyl, triazinyl, imidazo[1,2-b]pyrazolyl, furo[2,3- c]pyridinyl, imidazo[1,2-a]pyridinyl, indazolyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, pyrazolo[3,4-c]pyr
  • the aryl groups herein defined may have an unsaturated or partially saturated ring fused with a fully saturated ring.
  • exemplary ring systems of these heteroaryl groups include indolinyl, indolinonyl, dihydrobenzothiophenyl, dihydrobenzofuran, chromanyl, thiochromanyl, tetrahydroquinolinyl, dihydrobenzothiazine,3,4-dihydro- lH-isoquinolinyl, 2,3-dihydrobenzofuran, indolinyl, indolyl, and dihydrobenzoxanyl.
  • Halogen or“halo” mean fluorine, chlorine, bromine, or iodine.
  • Alkyl means a straight or branched chain saturated hydrocarbon containing 1-12 carbon atoms.
  • Examples of a (C 1 -C 6 )alkyl group include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, isopropyl, isobutyl, sec-butyl, tert-butyl, isopentyl, neopentyl, and isohexyl.
  • Alkoxy means a straight or branched chain saturated hydrocarbon containing 1-12 carbon atoms 25 containing a terminal“O” in the chain, e.g., -O(alkyl).
  • alkoxy groups include, without limitation, methoxy, ethoxy, propoxy, butoxy, t-butoxy, or pentoxy groups.
  • Alkenyl means a straight or branched chain unsaturated hydrocarbon containing 2-12 carbon atoms.
  • The“alkenyl” group contains at least one double bond in the chain.
  • the double bond of an alkenyl group can be unconjugated or conjugated to another unsaturated group.
  • alkenyl groups include 30 ethenyl, propenyl, n-butenyl, iso-butenyl, pentenyl, or hexenyl.
  • An alkenyl group can be unsubstituted or substituted and may be straight or branched.
  • Alkynyl means a straight or branched chain unsaturated hydrocarbon containing 2-12 carbon atoms.
  • The“alkynyl” group contains at least one triple bond in the chain.
  • alkenyl groups include ethynyl, propargyl, n-butynyl, iso-butynyl, pentynyl, or hexynyl.
  • An alkynyl group can be 35 unsubstituted or substituted.
  • Alkylene or“alkylenyl” means a divalent alkyl radical. Any of the above mentioned monovalent alkyl groups may be an alkylene by abstraction of a second hydrogen atom from the alkyl. As herein defined, alkylene may also be a (C 1 -C 6 )alkylene. An alkylene may further be a (C 1 -C 4 )alkylene.
  • Typical alkylene groups include, but are not limited to, -CH 2 -, -CH(CH 3 )-, -C(CH 3 ) 2 -, -CH 2 CH 2 -, -CH 2 CH(CH 3 )-, - CH 2 C(CH 3 ) 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH 2 CH 2 CH-, and the like.
  • Cycloalkyl or“carbocyclyl” means a monocyclic or polycyclic saturated or partially unsaturated 5 non-aromatic carbon ring containing 3-18 carbon atoms.
  • Examples of cycloalkyl groups include, without limitations, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptanyl, cyclooctanyl, norboranyl, norborenyl, bicyclo[2.2.2]octanyl, or bicyclo[2.2.2]octenyl and derivatives thereof.
  • a (C 3 -C 8 )cycloalkyl is a cycloalkyl group containing between 3 and 8 carbon atoms.
  • a cycloalkyl group can be fused (e.g., decalin) or bridged (e.g., norbornane).
  • Heterocyclyl or“heterocycloalkyl” means a saturated or partially saturated monocyclic or polycyclic ring containing carbon and at least one heteroatom selected from oxygen, nitrogen, or sulfur (O, N, or S) and wherein there is not delocalized n electrons (aromaticity) shared among the ring carbon or heteroatoms.
  • the heterocycloalkyl ring structure may be substituted by one or more substituents. The substituents can themselves be optionally substituted.
  • heterocyclyl rings include, but are not 15 limited to, oxetanyl, azetadinyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, oxazolinyl, oxazolidinyl, thiazolinyl, thiazolidinyl, pyranyl, thiopyranyl, tetrahydropyranyl, dioxalinyl, piperidinyl, morpholinyl, thiomorpholinyl, thiomorpholinyl S-oxide, thiomorpholinyl S-dioxide, piperazinyl, azepinyl, oxepinyl, diazepinyl, tropanyl, oxazolidinonyl, 1,4-dioxanyl, dihydrofuranyl, 1,3-dioxolanyl, imidazolidinyl, imidazolin
  • “Hydroxyalkyl” means an alkyl group substituted with one or more -OH groups. Examples of hydroxyalkyl groups include HO-CH 2 -, HO-CH 2 CH 2 -, and CH 2 -CH(OH)-.
  • Haloalkyl means an alkyl group substituted with one or more halogens.
  • haloalkyl groups include, but are not limited to, trifluoromethyl, difluoromethyl, pentafluoroethyl, trichloromethyl, etc.
  • Haloalkoxy means an alkoxy group substituted with one or more halogens.
  • haloalkyl groups include, but are not limited to, trifluoromethoxy, difluoromethoxy, pentafluoroethoxy, trichloromethoxy, etc.
  • Cyano means a substituent having a carbon atom joined to a nitrogen atom by a triple bond, e.g., CoN.
  • Amino means a substituent containing at least one nitrogen atom (e.g., NH2).
  • “Pomalidomide” or 4-amino-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione has the following structure:
  • Prodrug or“prodrug derivative” mean a covalently-bonded derivative or carrier of the parent compound or active drug substance which undergoes at least some biotransformation prior to exhibiting its pharmacological effect(s).
  • prodrugs have metabolically cleavable groups and are rapidly 5 transformed in vivo to yield the parent compound, for example, by hydrolysis in blood, and generally include esters and amide analogs of the parent compounds.
  • the prodrug is formulated with the objectives of improved chemical stability, improved patient acceptance and compliance, improved bioavailability, prolonged duration of action, improved organ selectivity, improved formulation (e.g., increased hydrosolubility), and/or decreased side effects (e.g., toxicity).
  • prodrugs themselves have weak 10 or no biological activity and are stable under ordinary conditions.
  • Prodrugs can be readily prepared from the parent compounds using methods known in the art, such as those described in A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard (eds.), Gordon & Breach, 1991, particularly Chapter 5:“Design and Applications of Prodrugs”; Design of Prodrugs, H. Bundgaard (ed.), Elsevier, 1985; Prodrugs: Topical and Ocular Drug Delivery, K.B. Sloan (ed.), Marcel Dekker, 1998; 15 Methods in Enzymology, K. Widder et al. (eds.), Vol.42, Academic Press, 1985, particularly pp.309-396;
  • “Pharmaceutically acceptable prodrug” as used herein means a prodrug of a compound of the disclosure which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible.
  • Salt means an ionic form of the parent compound or the product of the reaction between the parent compound with a suitable acid or base to make the acid salt or base salt of the parent compound.
  • Salts of the compounds of the present disclosure can be synthesized from the parent compounds which contain a basic or acidic moiety by conventional chemical methods. Generally, the salts are prepared by reacting the free base or acid parent compound with stoichiometric amounts or with an excess of the desired 30 salt-forming inorganic or organic acid or base in a suitable solvent or various combinations of solvents.
  • “Pharmaceutically acceptable salt” means a salt of a compound of the disclosure which is, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oil-soluble or dispersible, and effective for their intended use.
  • the term 35 includes pharmaceutically-acceptable acid addition salts and pharmaceutically-acceptable base addition salts.
  • the compounds of the present disclosure are useful in both free base and salt form, in practice, the use of the salt form amounts to use of the base form. Lists of suitable salts are found in, e.g., S.M. Birge et al., J. Pharm. Sci., 1977, 66, pp.1-19, which is hereby incorporated by reference in its entirety.
  • “Pharmaceutically-acceptable acid addition salt” means those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, 5 formed with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trichloroacetic acid, trifluoroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 2-acetoxybenzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, 10 hemisulfic acid, heptanoic acid, hexanoic acid, formic acid
  • “Pharmaceutically-acceptable base addition salt” means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a metal cation such as sodium, potassium, lithium, calcium, magnesium, iron, zinc, copper, manganese, 20 aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically-acceptable organic nontoxic bases include salts of primary, secondary, and tertiary amines, quaternary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion-exchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, 25 tributylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tetramethylammonium compounds, tetraethylammoni
  • Solvate means a complex of variable stoichiometry formed by a solute, for example, a compound of Formula (I)) and solvent, for example, water, ethanol, or acetic acid. This physical association may 35 involve varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. In general, such solvents selected for the purpose of the disclosure do not interfere with the biological activity of the solute. Solvates encompasses both solution- phase and isolatable solvates. Representative solvates include hydrates, ethanolates, methanolates, and the like.
  • “Hydrate” means a solvate wherein the solvent molecule(s) is/are water.
  • the compounds of the present disclosure as discussed below include the free base or acid thereof, their salts, solvates, and prodrugs and may include oxidized sulfur atoms or quaternized nitrogen atoms in their structure, although not explicitly stated or shown, particularly the pharmaceutically acceptable forms thereof. Such forms, particularly the pharmaceutically acceptable forms, are intended to be embraced by the appended claims.
  • “Isomers” means compounds having the same number and kind of atoms, and hence the same molecular weight, but differing with respect to the arrangement or configuration of the atoms in space.
  • the term includes stereoisomers and geometric isomers.
  • Stepoisomer or“optical isomer” mean a stable isomer that has at least one chiral atom or 15 restricted rotation giving rise to perpendicular dissymmetric planes (e.g., certain biphenyls, allenes, and spiro compounds) and can rotate plane-polarized light. Because asymmetric centers and other chemical structure exist in the compounds of the disclosure which may give rise to stereoisomerism, the disclosure contemplates stereoisomers and mixtures thereof.
  • the compounds of the disclosure and their salts include asymmetric carbon atoms and may therefore exist as single stereoisomers, racemates, and as mixtures of 20 enantiomers and diastereomers. Typically, such compounds will be prepared as a racemic mixture.
  • stereoisomers can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures.
  • individual stereoisomers of compounds are prepared by synthesis from optically active starting materials containing the desired chiral centers or by preparation of mixtures of enantiomeric products followed by 25 separation or resolution, such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, use of chiral resolving agents, or direct separation of the enantiomers on chiral chromatographic columns.
  • Starting compounds of particular stereochemistry are either commercially available or are made by the methods described below and resolved by techniques well-known in the art.
  • Enantiomers means a pair of stereoisomers that are non-superimposable mirror images of each other.
  • Racemic mixture or“racemate” mean a mixture containing equal parts of individual enantiomers.
  • the substituents 5 and the isomers are designated using the cis/trans convention or using the E or Z system, wherein the term “E” means higher order substituents on opposite sides of the double bond, and the term“Z” means higher order substituents on the same side of the double bond.
  • E means higher order substituents on opposite sides of the double bond
  • Z means higher order substituents on the same side of the double bond.
  • Some of the compounds of the disclosure can exist in more than one tautomeric form. As mentioned above, the compounds of the disclosure include all such tautomers.
  • enantiomers often exhibit strikingly different biological activity including differences in pharmacokinetic properties, including metabolism, protein binding, and the like, and pharmacological properties, including the type of activity displayed, the degree of activity, toxicity, and the like.
  • one enantiomer may be more active or may exhibit beneficial effects when enriched relative to the other enantiomer or when 20 separated from the other enantiomer.
  • one skilled in the art would know how to separate, enrich, or selectively prepare the enantiomers of the compounds of the disclosure from this disclosure and the knowledge of the prior art.
  • racemic form of drug may be used, it is often less effective than administering an equal amount of enantiomerically pure drug; indeed, in some cases, one enantiomer may be 25 pharmacologically inactive and would merely serve as a simple diluent.
  • ibuprofen had been previously administered as a racemate, it has been shown that only the S-isomer of ibuprofen is effective as an anti-inflammatory agent (in the case of ibuprofen, however, although the R-isomer is inactive, it is converted in vivo to the S-isomer, thus, the rapidity of action of the racemic form of the drug is less than that of the pure S-isomer).
  • enantiomers may have 30 distinct biological activity.
  • S-penicillamine is a therapeutic agent for chronic arthritis, while R-penicillamine is toxic.
  • R-penicillamine is toxic.
  • some purified enantiomers have advantages over the racemates, as it has been reported that purified individual isomers have faster transdermal penetration rates compared to the racemic mixture. See U.S. Pat. Nos.5,114,946 and 4,818,541.
  • one enantiomer is pharmacologically more active, less toxic, or has a preferred disposition 35 in the body than the other enantiomer, it would be therapeutically more beneficial to administer that enantiomer preferentially. In this way, the patient undergoing treatment would be exposed to a lower total dose of the drug and to a lower dose of an enantiomer that is possibly toxic or an inhibitor of the other enantiomer.
  • Preparation of pure enantiomers or mixtures of desired enantiomeric excess (ee) or enantiomeric purity are accomplished by one or more of the many methods of (a) separation or resolution of enantiomers, 5 or (b) enantioselective synthesis known to those of skill in the art, or a combination thereof.
  • These resolution methods generally rely on chiral recognition and include, for example, chromatography using chiral stationary phases, enantioselective host-guest complexation, resolution or synthesis using chiral auxiliaries, enantioselective synthesis, enzymatic and nonenzymatic kinetic resolution, or spontaneous enantioselective crystallization.
  • A“patient” or“subject” is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, or nonhuman primate, such as a monkey, chimpanzee, baboon or, rhesus.
  • the subject is a primate.
  • the subject is a human.
  • an“effective amount” or“therapeutically effective amount” when used in connection with a compound means an amount of a compound of the present disclosure that (i) treats or prevents the particular 25 disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • “pharmaceutically effective amount” or“therapeutically effective amount” means an amount of a compound according to the disclosure which, when administered to a patient in need thereof, 30 is sufficient to effect treatment for disease-states, conditions, or disorders for which the compounds have utility. Such an amount would be sufficient to elicit the biological or medical response of a tissue, system, or patient that is sought by a researcher or clinician.
  • the amount of a compound of according to the disclosure which constitutes a therapeutically effective amount will vary depending on such factors as the compound and its biological activity, the composition used for administration, the time of administration, 35 the route of administration, the rate of excretion of the compound, the duration of treatment, the type of disease-state or disorder being treated and its severity, drugs used in combination with or coincidentally with the compounds of the disclosure, and the age, body weight, general health, sex, and diet of the patient.
  • a therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their own knowledge, the prior art, and this disclosure.
  • the term“pharmaceutical composition” refers to a compound of the disclosure, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, together 5 with at least one pharmaceutically acceptable carrier, in a form suitable for oral or parenteral administration.
  • Carrier encompasses carriers, excipients, and diluents and means a material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a pharmaceutical agent from one organ, or portion of the body, to another organ, or portion of the body of a subject.
  • a subject is“in need of” a treatment if such subject would benefit biologically, medically, or in quality of life from such treatment (preferably, a human).
  • the term“inhibit”,“inhibition”, or“inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term“treat”,“treating”, or “treatment” of any disease or disorder refers to alleviating or ameliorating the disease or disorder (i.e., slowing or arresting the development of the disease or at least one of the clinical symptoms thereof); or alleviating or ameliorating at least one physical parameter or biomarker associated with the disease or disorder, including those which may not be discernible to the patient.
  • the term“prevent”,“preventing”, or“prevention” of any disease or disorder refers to the prophylactic treatment of the disease or disorder; or delaying the onset or progression of the disease or disorder.
  • “Pharmaceutically acceptable” means that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal 25 being treated therewith.
  • disorder means, and is used interchangeably with, the terms disease, condition, or illness, unless otherwise indicated.
  • administering means to either directly administering a disclosed compound or pharmaceutically acceptable salt of the disclosed compound or a composition to a 30 subject, or administering a prodrug derivative or analog of the compound or pharmaceutically acceptable salt of the compound or composition to the subject, which can form an equivalent amount of active compound within the subject’s body.
  • Prodrug means a compound which is convertible in vivo by metabolic means (e.g., by hydrolysis) to a disclosed compound.
  • Compounds of the present disclosure refer to compounds of Formulae (I), (Ia), (Ib), (Ic), (Id), (Ie), (If), (Ig), (Ih), (Ii), (Ij), (Ik), and (Il), as herein described including the tautomers, the prodrugs, salts particularly the pharmaceutically acceptable salts, and the solvates and hydrates thereof, where the context so permits thereof, as well as all stereoisomers (including diastereoisomers and enantiomers), rotamers, tautomers, and isotopically labelled compounds (including deuterium substitutions), as well as inherently formed moieties (e.g., polymorphs, solvates and/or hydrates).
  • solvates and hydrates are 5 generally considered compositions.
  • the compounds of the disclosure and the formulas designating the compounds of the disclosure are understood to only include the stable compounds thereof and exclude unstable compounds, even if an unstable compound might be considered to be literally embraced by the compound formula.
  • reference to intermediates, whether or not they themselves are claimed, is meant to embrace their salts and solvates, where the context so permits.
  • particular instances when the context so permits are sometimes indicated in the text, but these instances are purely illustrative and it is not intended to exclude other instances when the context so permits.
  • “Stable compound” or“stable structure” means a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic or diagnostic agent.
  • a compound, which would have a“dangling valency” or is a carbanion 15 is not a compound contemplated by the disclosure.
  • the term“about” or“approximately” means within 20%, preferably within 10%, and more preferably within 5% of a given value or range.
  • Cancer means any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, 20 neoplasms, carcinomas, sarcomas, leukemias, lymphomas, and the like.
  • cancers include, but are not limited to, mesothelioma, leukemias, and lymphomas such as cutaneous T-cell lymphomas (CTCL), noncutaneous peripheral T-cell lymphomas, lymphomas associated with human T-cell lymphotrophic virus (HTLV) such as adult T-cell leukemia/lymphoma (ATLL), B-cell lymphoma, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, acute myelogenous leukemia, 25 lymphomas, and multiple myeloma, non-Hodgkin lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), Hodgkin’s lymphoma, Burkitt lymphoma, adult T-cell leukemia lymphoma, acute-myeloid leukemia (AML), chronic myeloid leukemia (CML), or hepatocellular carcinoma.
  • CCL cutaneous T-cell lymph
  • myelodisplastic syndrome childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms’ tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such 30 as head and neck cancers (e.g., oral, laryngeal, and nasopharyngeal), esophageal cancer, genitourinary cancers (e.g., prostate, bladder, renal, uterine, ovarian, testicular), lung cancer (e.g., small-cell and non- small cell), breast cancer, pancreatic cancer, melanoma, and other skin cancers, stomach cancer, brain tumors, tumors related to Gorlin’s syndrome (e.g., medulloblastoma, meningioma, etc.), and liver cancer.
  • childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms’ tumor, bone tumors,
  • Additional exemplary forms of cancer which may be treated by the subject compounds include, but are not 35 limited to, cancer of skeletal or smooth muscle, stomach cancer, cancer of the small intestine, rectum carcinoma, cancer of the salivary gland, endometrial cancer, adrenal cancer, anal cancer, rectal cancer, parathyroid cancer, and pituitary cancer. Additional cancers that the compounds described herein may be useful in preventing, treating, and studying are, for example, colon carcinoma, familiary adenomatous polyposis carcinoma, and hereditary non-polyposis colorectal cancer, or melanoma.
  • cancers include, but are not limited to, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, 5 gastric carcinoma, adenocarcinoma, thyroid cancer (medullary and papillary thyroid carcinoma), renal carcinoma, kidney parenchyma carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, testis carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, gall bladder carcinoma, bronchial carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, 10 choroidea melanoma, seminoma, rhabdomyosarcoma, craniopharyngeoma, osteosarcoma, chondrosarcoma, myosarcom
  • “Simultaneously” or“simultaneous” when referring to a method of treating or a therapeutic use means with a combination of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and one or more second agent(s) means administration 15 of the compound and the one or more second agent(s) by the same route and at the same time.
  • “Separately” or“separate” when referring to a method of treating or a therapeutic use means with a combination of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and one or more second agent(s) means administration of the compound and the one or more second agent(s) by different routes and at approximately the same time.
  • therapeutic administration“over a period of time” means, when referring to a method of treating or a therapeutic use with a combination of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and one or more second agent(s), administration of the compound and the one or more second agent(s) by the same or different routes and at different times.
  • the administration of the compound or the one or more second 25 agent(s) occurs before the administration of the other begins.
  • a one of the active ingredients i.e., a compound of the Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or one or more second agent(s)
  • Another therapeutic administration over a period of time consists of the administration over time of 30 the two or more active ingredients of the combination using different frequencies of administration for each of the active ingredients, whereby at certain time points in time simultaneous administration of all of the active ingredients takes place whereas at other time points in time only a part of the active ingredients of the combination may be administered (e.g., for example.
  • IKZF2-dependent disease or disorder means any disease or disorder which is directly or indirectly affected by the modulation of IKZF2 protein levels.
  • IKZF4-dependent disease or disorder means any disease or disorder which is directly or indirectly affected by the modulation of IKZF4 protein levels.
  • the present disclosure relates to compounds or pharmaceutically acceptable salts, hydrates, solvates, prodrugs, stereoisomers, or tautomers thereof, capable of modulating IKZF2 protein levels, which are useful for the treatment of diseases and disorders associated with modulation of IKZF2 protein levels.
  • the disclosure further relates to compounds, or pharmaceutically acceptable salts, hydrates, solvates, 10 prodrugs, stereoisomers, or tautomers thereof, which are useful for reducing or decreasing IKZF2 protein levels.
  • the compounds of Formula (I) have the structure of Formula (Ia):
  • the compounds of Formula (I) have the structure of Formula (Ib):
  • the compounds of Formula (I) have the structure of Formula (Ic):
  • the compounds of Formula (I) have the structure of Formula (Id):
  • the compounds of Formula (I) have the structure of Formula (Ie):
  • the compounds of Formula (I) have the structure of Formula (If):
  • the compounds of Formula (I) have the structure of Formula (Ig):
  • the compounds of Formula (I) have the structure of Formula (Ih):
  • the compounds of Formula (I) have the structure of Formula (Ii):
  • the compounds of Formula (I) have the structure of Formula (Ij):
  • the compounds of Formula (I) have the structure of Formula (Ik):
  • the compounds of Formula (I) have the structure of Formula (Il):
  • R 2 is (C 1 -C 6 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the alkyl is optionally substituted with one to four R 4 ; and the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to four R 5 , or 10 R 1 and R 2 , when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6- membered heterocycloalkyl ring;
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, -OH, - NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 4- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms 15 selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 ;
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 - C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 3 - C 7 )cycloalkyl, 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and 20 S, (C 6 -C 10 )aryl, and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, or
  • R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 - C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 , or
  • R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 5 - C 7 )cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to four R 10 ;
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 - C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -(CH 2 ) 0-3 C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 ,30 -NR 8 C(O)OR 9 , -S(O) p NR 8 R 9 , -S(O) p R 12 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, - O(CH 2 ) 0-3 (C 6 -C 10 )aryl, adam
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, (C 6 -C 10 )aryl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and 10 heterocycloalkyl are optionally substituted with one to four substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, - NH 2 , and CN;
  • R x is D. In another embodiment, R x is H.
  • X 1 is CR 3 .
  • X 2 is N and X 3 is CR 14 .
  • X 2 is CR 13 and X 3 is N.
  • X 2 is CR 15 and X 3 is CR 14 .
  • X 2 is CR 13 and X 3 is CR 16 .
  • X 2 is N and X 3 is CH.
  • X 2 is CH and X 3 is 20 N.
  • X 2 is CH and X 3 is CR 16 .
  • X 2 is CR 15 and X 3 is CH.
  • each R 1 is independently (C 1 -C 6 )haloalkyl, (C 1 - C 6 )hydroxyalkyl, CN, or halogen. In another embodiment, each R 1 is independently (C 1 -C 6 )alkyl, (C 1 - C 6 )haloalkyl, CN, or halogen. In yet another embodiment, each R 1 is independently (C 1 -C 6 )alkyl, (C 1 - C 6 )hydroxyalkyl, CN, or halogen. In another embodiment, each R 1 is independently (C 1 -C 6 )alkyl, (C 1 -25 C 6 )haloalkyl, CN, or halogen. In yet another embodiment, each R 1 is independently (C 1 -C 6 )alkyl or (C 1 - C 6 )haloalkyl.
  • each R 1 is independently (C 1 -C 6 )haloalkyl, (C 1 -C 6 )hydroxyalkyl, or halogen. In another embodiment, each R 1 is independently (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, or halogen. In yet another embodiment, each R 1 is independently (C 1 -C 6 )alkyl, (C 1 -C 6 )hydroxyalkyl, or halogen. In 30 another embodiment, each R1 is independently (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, or halogen.
  • each R 1 is independently (C 1 -C 6 )alkyl or (C 1 -C 6 )haloalkyl. In another embodiment, each R 1 is independently (C 1 -C 6 )alkyl or halogen. In yet another embodiment, each R 1 is independently (C 1 - C 6 )haloalkyl or halogen. In another embodiment, each R 1 is independently D or (C 1 -C 6 )alkyl. In another embodiment, each R 1 is independently (C 1 -C 6 )alkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 3 -C 7 )cycloalkyl or a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a (C 3 -C 7 )cycloalkyl or a 5- or 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a (C3-C7)cycloalkyl or a 4- or 5- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the 5 carbon atoms to which they are attached form a (C 4 -C 7 )cycloalkyl or a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a (C 4 -C 6 )cycloalkyl or a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a 10 (C3-C7)cycloalkyl.
  • two R1 together with the carbon atoms to which they are attached form a (C 3 -C 6 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 4 -C 7 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 5 -C 7 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 6 -C 7 )cycloalkyl.
  • two R 1 15 together with the carbon atoms to which they are attached form a (C 5 -C 6 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 4 -C 6 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 3 -C 6 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached form a (C 3 - C 5 )cycloalkyl.
  • two R 1 together with the carbon atoms to which they are attached 20 form a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a 5- or 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 together with the carbon atoms to which they are attached form a 4- or 5- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 when on adjacent atoms, together with the atoms to which they are attached form a phenyl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 when on adjacent atoms, together with the atoms to which they are attached form a phenyl ring.
  • two R 1 when on adjacent atoms, together with the atoms to which they are attached form a phenyl ring.
  • two R1 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 when on adjacent atoms, together with the atoms to which they are attached form a 5-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • two R 1 when on adjacent atoms, together with the atoms to which they are attached form a 35 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S.
  • R 2 is (C 1 -C 6 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the alkyl is optionally substituted with one to four R 4 ; and the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to four R5.
  • R 2 is (C 1 -C 4 )alkyl, (C6-C10)aryl, ( (C 3 -C) 8 cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein 5 the alkyl is optionally substituted with one to three R 4 ; and wherein the aryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 1 -C 4 )alkyl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the alkyl is optionally substituted with one to three R 4 ; and wherein the heteroaryl, cycloalkyl, and 10 heterocycloalkyl are optionally substituted with one to three R5.
  • R 2 is (C 1 -C 4 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, or (C 3 - C 8 )cycloalkyl, wherein the alkyl is optionally substituted with one to three R 4 ; and wherein the aryl, heteroaryl, and cycloalkyl, are optionally substituted with one to three R 5 .
  • R 2 is (C 1 -C 4 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, 15 and S, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the alkyl is optionally substituted with one to three R 4 ; and wherein the aryl, heteroaryl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl 20 comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is phenyl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl 25 comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the phenyl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 1 -C 3 )alkyl optionally substituted with one to three R 4 .
  • R 2 is (C 1 -C 3 )alkyl substituted with one to three R 4 .
  • R 2 is (C 3 -C 8 )cycloalkyl or 5- to 7-membered heterocycloalkyl comprising 30 1 to 3 heteroatoms selected from O, N, and S, wherein the cycloalkyl and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 6 -C 10 )aryl or 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl are optionally substituted with one to three R 5 .
  • R 2 is (C 3 -C 8 )cycloalkyl or (C 6 -C 10 )aryl, wherein the cycloalkyl and aryl are optionally substituted with one to three R 5 .
  • 35 R 2 is 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, or 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the heteroaryl and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 6 -C 10 )aryl optionally substituted with one to three R 5 .
  • R 2 is 5- or 6- membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R5.
  • R 2 is (C3-C8)cycloalkyl optionally substituted with one to three R5.
  • R 2 is 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 5 .
  • R 1 and R 2 when on adjacent atoms, together with the atoms to which they are attached form a 5-membered heterocycloalkyl ring. In another embodiment, R 1 and R 2 , when on adjacent atoms, together with the atoms to which they are attached form a 6-membered heterocycloalkyl ring.
  • R3 is D. In another embodiment, R3 is H. In another embodiment, R 3 is absent when s a double bond.
  • each R 4 is independently selected from -C(O)OR 6 , - C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, -OH, -NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 4- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 .
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, -OH, -NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 .
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , - NR 6 C(O)R 6' , halogen,–OH, -NH 2 , or CN. In another embodiment, each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, or -OH.
  • each R 4 is independently selected from halogen, -OH, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 .
  • each R 4 is independently selected from halogen, -OH, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 .
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , and - NR 6 C(O)R 6' .
  • each R 4 is independently selected from -C(O)OR 6 , (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R 7 .
  • each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to four R7.
  • each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 5 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • each R 4 is independently selected from (C 6 -C 10 )aryl and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl are 10 optionally substituted with one to three R7.
  • each R4 is independently selected from (C 6 -C 10 )aryl and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl are substituted with one to three R 7 .
  • each R 4 is independently selected from (C 3 -C 8 )cycloalkyl and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the 15 cycloalkyl and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • each R 4 is independently selected from (C 3 -C 8 )cycloalkyl and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the cycloalkyl and heterocycloalkyl groups are substituted with one to three R 7 .
  • each R 4 is independently (C 6 -C 10 )aryl optionally substituted with one to 20 three R 7 .
  • each R 4 is independently 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 7 .
  • each R 4 is (C 3 -C 8 )cycloalkyl optionally substituted with one to three R 7 .
  • each R 4 is independently 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 7 .
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 3 -C 7 )cycloalkyl, 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 6 -C 10 )aryl, and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • each R 5 is independently selected from30 (C 1 -C 6 )alkyl, (C2-C6)alkenyl, (C2-C6)alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C1- C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 5 is independently selected from (C 3 -C 7 )cycloalkyl, 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 6 -C 10 )aryl, and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 - C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 3 -C 7 )cycloalkyl, 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 6 -C 10 )aryl, and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • each R5 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C1- C 6 )haloalkyl, and (C 1 -C 6 )haloalkoxy.
  • each R 5 is independently selected from 5 (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, and CN.
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring 10 comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R10, or two R 5 , when on adjacent atoms, together with the atoms to which they are attached form a (C 5 - C 7 )cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to four R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6- 15 membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally
  • R 10 substituted with one to three R 10 , or two R 5 , when on adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 7 )cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they 20 are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 7 )cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring optionally substituted with one to three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a phenyl ring optionally substituted with one to three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6-membered heteroaryl ring comprising 1 30 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R10.
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 7 )cycloalkyl ring optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 7 )cycloalkyl ring optionally substituted with one three R 10 .
  • two R 5 when on 35 adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 6 )cycloalkyl ring
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 5 )cycloalkyl ring optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 )cycloalkyl ring optionally substituted with one three R 10 .
  • two R5, when on adjacent atoms, together with the atoms to which they are attached form a (C 7 )cycloalkyl ring optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R5 when on adjacent atoms, together with the atoms to which they are attached form a 6- or 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 5-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 6-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • two R 5 when on adjacent atoms, together with the atoms to which they are attached form a 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one three R 10 .
  • R 6 is H or (C 1 -C 3 )alkyl. In another embodiment, R 6 is H or (C 6 -C 10 )aryl. In yet another embodiment, R 6 is (C 1 -C 3 )alkyl or (C 6 -C 10 )aryl. In another embodiment, R 6 is H, methyl, ethyl, n-propyl, or isopropyl. In another embodiment, R 6 is H, methyl or ethyl. In yet another embodiment, R 6 is H or methyl. In another embodiment, R 6 is H.
  • R 6' is H or (C 1 -C 3 )alkyl.
  • R 6' 25 is H or (C 6 -C 10 )aryl.
  • R 6' is (C 1 -C 3 )alkyl or (C 6 -C 10 )aryl.
  • R 6' is H, methyl, ethyl, n-propyl, or isopropyl. In another embodiment, R 6' is H, methyl or ethyl. In yet another embodiment, R 6' is H or methyl. In another embodiment, R 6' is H.
  • each R 7 is independently selected from (C 1 - C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -30 (CH 2 )0-3C(O)OR8, -C(O)NR8R9, -NR8C(O)R9, -NR8C(O)OR9, -S(O)pNR8R9, -S(O)pR12, (C1- C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, -O(CH 2 ) 0-3 (C 6 -C 10 )aryl, adamantyl,
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 - C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -(CH 2 ) 0-3 C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , -NR 8 C(O)OR 9 , -S(O)pNR8R9, -S(O)pR12, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 )1-3CN, -NH2, CN, -O(CH 2 )0-3(C6- C 10 )aryl, -O(CH 2 ) 0-3
  • each R7 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C1- C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -(CH 2 ) 0-3 C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , -NR 8 C(O)OR 9 , -S(O) p NR 8 R 9 , -S(O) p R 12 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, -O(CH 2 ) 0-3 (C 6 - C 10 )aryl, -O(CH 2 ) 0-3 -5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms
  • each R 7 is independently selected from -(CH 2 ) 0-3 C(O)OR 8 , 20 -NR 8 C(O)OR 9 , -S(O) p NR 8 R 9 , -S(O) p R 12 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, -O(CH 2 ) 0-3 (C 6 -C 10 )aryl, -O(CH 2 ) 0-3 -5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, bicyclic 9- or 10-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl and heterocycloalkyl are optionally substituted with one to four substituents each independently selected from halogen, (C 1 -C 6 )alkyl,
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 - C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -C(O)OR 8 , -C(O)NR 8 R 9 , - NR 8 C(O)R 9 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 7 )cycloalkyl, and 5- to 7-membered 30 heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 - C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 - 35 C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 7 is independently selected from (C 1 - C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy.
  • each R 7 is independently selected from -C(O)R 8 , -C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 7 is independently selected from (C 6 - C10)aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C3- C 7 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, 5 and S.
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 - C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 7 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 10 heteroatoms selected from O, N, and S.
  • each R7 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 .
  • two R 7 together with the atoms to which they are attached form a (C 5 - 20 C 7 ) cycloalkyl ring optionally substituted with one to four R 10 .
  • two R 7 together with the atoms to which they are attached form a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms 25 selected from O, N, and S, optionally substituted with one to four R 10 , or two R 7 , when on adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 7 )cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they 30 are attached form a (C5-C7)cycloalkyl ring or a 5- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 5 -C 7 )cycloalkyl ring optionally substituted with one to four R 10 .
  • two R 7 when on adjacent atoms, together with the atoms to which they are attached form a 5- to 7-membered
  • R 8 is H or (C 1 -C 3 )alkyl.
  • R 8 is H, methyl, ethyl, n-propyl, or isopropyl.
  • R 8 is H, methyl or ethyl.
  • R8 is H or methyl.
  • R8 is H
  • R 9 is H or (C 1 -C 3 )alkyl.
  • R 9 5 is H, methyl, ethyl, n-propyl, or isopropyl.
  • R 9 is H, methyl or ethyl.
  • R 9 is H or methyl.
  • R 9 is H.
  • each R 10 is independently selected from (C 1 - C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, and halogen.
  • each R 10 is independently selected from -OH, -NH 2 , and CN.
  • each R10 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C1- C 6 )haloalkoxy, and halogen.
  • each R 10 is independently selected from (C 1 - C 6 )alkyl, (C 1 -C 6 )haloalkyl, and halogen. In yet another embodiment, each R 10 is independently selected from (C 1 -C 6 )alkyl and halogen.
  • each R 11 is independently selected from CN, (C 1 - C 6 )alkoxy, (C 6 -C 10 )aryl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heterocycloalkyl are optionally substituted with one to four substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 - 20 C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, (C 6 -C 10 )aryl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heterocycloalkyl are optionally substituted with one to three substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 - C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , and CN.
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, and (C 6 -C 10 )aryl, wherein the aryl is optionally substituted with one to three substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, - NH 2 , and CN.
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, and 5- to 7- 30 membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the heterocycloalkyl is optionally substituted with one to four substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, - NH 2 , and CN.
  • each R 11 is independently selected from CN and (C 1 -C 6 )alkoxy. In yet another embodiment, each R 11 is independently selected from (C 6 -C 10 )aryl and 5- to 7-membered 35 heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and
  • heterocycloalkyl are optionally substituted with one to four substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, - NH 2 , and CN.
  • R12 is (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C6-C10)aryl, or 5- or 6-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • R 12 is (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, phenyl, or 5- or 6-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • R 12 is (C 1 -C 4 )alkyl, (C 1 -C 4 )haloalkyl, phenyl, or 5- or 6-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • R 13 is halogen, -OH, or -NH 2 .
  • R13 is H, halogen, or -NH2.
  • R13 is H, F, Cl, or -NH2.
  • R 13 is H, F, Cl, -OH, or-NH 2 .
  • R 13 is H, F, or -NH 2 .
  • R 13 is F or -NH 2 .
  • R 14 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 - C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 - C 3 )hydroxyalkyl, F, Cl, -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 - C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )hydroxyalkyl, F, Cl, -OH, - NH 2 , -NO 2 , or CN.
  • R 14 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, halogen,20 -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 - C 3 )haloalkyl, F, Cl, -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 - C 3 )alkoxy, (C 1 -C 3 )haloalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN.
  • R 14 is H, (C 1 - C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, F, Cl, -OH, -NH 2 , -NO 2 , or CN.
  • R 15 is halogen, -OH, or -NH 2 .
  • R 15 is F, Cl, or -NH 2 .
  • R 15 is F, Cl, -OH, or -NH 2 .
  • R 15 is F, Cl, -OH, or -NH 2 .
  • R 15 is F or -NH 2 .
  • R 16 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 - C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN.
  • R 16 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -30 NH2, -NO2, or CN.
  • R16 is H, (C1-C3)alkyl, (C1-C3)alkoxy, (C1-C3)haloalkyl, (C1- C 3 )hydroxyalkyl, F, Cl, -OH, -NH 2 , -NO 2 , or CN.
  • R 16 is (C 1 -C 3 )alkyl, (C 1 - C 3 )alkoxy, (C 1 -C 3 )haloalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN.
  • R 16 is (C 1 - C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, F, Cl, -OH, -NH 2 , -NO 2 , or CN.
  • p is 0 or 1. In another embodiment, p is 1 or 2. In 35 yet another embodiment, p is 0 or 2. In another embodiment, p is 0. In yet another embodiment, p is 1. In another embodiment, p is 2. In some embodiments of the formulae above, n is 0 or 1. In another embodiment, n is 1 or 2. In yet another embodiment, n is 0 or 2. In another embodiment, n is 0. In yet another embodiment, n is 1. In another embodiment, n is 2.
  • n1 is 1. In another embodiment, n1 is 2.
  • n is 0 and n1 is 1. In another embodiment, n is 1 and n1 is 2. In another embodiment, n is 2 and n1 is 1. In another embodiment, n is 1 and n1 is 1.
  • q is 0, 1, 2, or 3. In another embodiment, q is 1, 2, 3, or 4. In yet another embodiment, q is 0, 1, or 2. In another embodiment, q is 1, 2, or 3. In yet another 10 embodiment, q is 2, 3, or 4. In another embodiment, q is 0 or 1. In yet another embodiment, q is 1 or 2. In another embodiment, q is 2 or 3. In yet another embodiment, q is 3 or 4. In another embodiment, q is 0. In yet another embodiment, q is 1. In another embodiment, q is 2. In yet another embodiment, q is 3. In another embodiment, q is 4.
  • X 1 is CH and n is 1. In another embodiment, X 1 is 15 CH, n is 1, and q is 0.
  • X 1 is CH, X 2 is N, and n is 1. In another embodiment, X 1 is CH, X 2 is N, n is 1, and q is 0.
  • X 1 is CH, X 3 is N, and n is 1. In another embodiment, X 1 is CH, X 3 is N, n is 1, and q is 0.
  • X 1 is CH, X 2 is N, and n is 1. In another embodiment, X 1 is CH, X 2 is N, n is 1, and q is 0, 1, or 2.
  • X 1 is CH, X 3 is N, and n is 1. In another embodiment, X 1 is CH, X 3 is N, n is 1, and q is 0, 1, or 2.
  • X 1 is CH, n is 1, and q is 0 or 1.
  • X 1 is CH, n is 1, q is 0 or 1, and R 1 is (C 1 -C 6 )alkyl.
  • X 1 is CH, n is 1, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with 30 one to three R4.
  • X1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is N, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 3 is N, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 13 , n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R4.
  • X 1 is CH, X 3 is CR 14 , n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally 5 substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 15 , n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X1 is CH
  • X3 is CR16
  • n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH
  • n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 13 , X 3 is CR 16 , n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 - 15 C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH
  • X 2 is CR 14
  • X 3 is CR 15
  • n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH
  • n is 1, q is 0, and R 2 is (C 1 - C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is20 (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is independently selected from - C(O)OR 6 , (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is independently selected from -C(O)OR 6 , (C 6 - C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with 30 one to three R7.
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is independently selected from (C 6 - C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, 35 and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C3-C8)cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the 5 aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • X 1 is CH
  • n is 1
  • q is 0, and R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7- 10 membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 6 -C 10 )aryl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0, and R 2 is 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 3 -C 8 )cycloalkyl 15 optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0, and R 2 is 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms 20 selected from O, N, and S, wherein the aryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • X 1 is CH, n is 1, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl, and 25
  • R 2 is (C 6 -C 10 )aryl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0, and R 2 is 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 3 -C 8 )cycloalkyl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is 5- to 7-membered heterocycloalkyl comprising 1 to 3 30 heteroatoms selected from O, N, and S, optionally substituted with one to three R5.
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, and R 2 is (C 1 - C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally 35 substituted with one to three R 4 , and each R 4 is independently selected from -C(O)OR 6 , (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from -C(O)OR 6 , (C 6 -C 10 )aryl, 5- or 6-membered 5 heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, (C 6 -C 10 )aryl, 5- 10 or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C3-C8)cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, (C 6 -C 10 )aryl, 5- or 6- 15 membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 0,
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, (C 6 -20 C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 0,
  • R 2 is (C 1 -C 6 )alkyl 25 substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0,
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, phenyl, 5- or 6- 5 membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, phenyl, 5- or 6-membered10 heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C3-C8)cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, n1 is 1, q is 0,
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH,15 phenyl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 0,
  • R 2 is (C 1 -C 6 )alkyl20 substituted with one to three R 4 , and each R 4 is independently selected from halogen, -OH, phenyl, 5- or 6- membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally 25 substituted with one to three R 4 , and each R 4 is independently selected from phenyl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl substituted 30 with one to three R4, and each R4 is independently selected from phenyl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 0,
  • R 2 is (C 1 -C 6 )alkyl35 optionally substituted with one to three R 4
  • each R 4 is independently selected from phenyl, 5- or 6- membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X1 is CH, n is 1, n1 is 1, q is 0,
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from phenyl, 5- or 6-membered 5 heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 , and each R 4 is independently selected from phenyl and 5- or 6-membered 10 heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 1, q is 0, R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from phenyl and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered 15 heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heteroaryl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is independently selected from phenyl and 5- or 6- membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and 20 heteroaryl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • n1 is 1
  • q is 0,
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 , and each R 4 is independently selected from phenyl and 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and 25 heteroaryl groups are optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is phenyl optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is phenyl optionally substituted with one to three R 7 .
  • X1 is CH
  • n is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is phenyl optionally substituted with one to three R 7 .
  • X 1 is CH
  • n is 1
  • q is 1
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is phenyl optionally substituted with one to three R 7 .
  • X 1 is CH and n is 2. In another embodiment, X 1 is CH, n is 2, and q is 0. In yet another embodiment, X 1 is CH, n is 2, and q is 0 or 1. In another embodiment, X 1 is CH, n is 2, q is 0 or 1, and R 1 is (C 1 -C 6 )alkyl. In some embodiments of the formulae above, X 1 is CH, n is 2, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 . In another embodiment, X 1 is CH, n is 2, q is 0 or 1, R1 is (C 1 -C 6 )alkyl, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R4.
  • X 1 is CH, n is 2, q is 0, and R 2 is (C 1 -C 6 )alkyl 5 optionally substituted with one to three R 4 .
  • X 1 is CH, n is 2, q is 0, and R 2 is (C 1 - C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 2, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is independently selected from - C(O)OR 6 , (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, 10 and S, (C3-C8)cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 2, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is independently selected from -C(O)OR 6 , (C 6 -15 C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 2, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is20 (C 1 -C 6 )alkyl optionally substituted with one to three R 4
  • each R 4 is independently selected from (C 6 - C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 - C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .
  • X 1 is CH, n is 2, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4
  • each R 4 is independently selected from (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one to three R 7 .30
  • X1 is CH, n is 2, q is 0, and R 2 is (C6-C10)aryl, (C3- C 8 )cycloalkyl, or 5- to
  • X 1 is CH, n is 2, q is 0, and R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7- membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • X 1 is CH, n is 2, q is 0, and R 2 is (C 6 -C 10 )aryl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0, and R 2 is 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0, and R 2 is (C 3 -C 8 )cycloalkyl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0, and R 2 is 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0 or 1
  • R 1 is (C 1 -C 6 )alkyl
  • R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 6 -C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms 10 selected from O, N, and S.
  • X 1 is CH, n is 2, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is (C 6 -C 10 )aryl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0, and R 2 is 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, 15 and R 2 is (C 3 -C 8 )cycloalkyl optionally substituted with one to three R 5 .
  • X 1 is CH, n is 2, q is 0 or 1, R 1 is (C 1 -C 6 )alkyl, and R 2 is 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one to three R 5 .
  • X 1 is CH, n is 1, n1 is 1, and R 2 is (C 1 -C 6 )alkyl optionally substituted with one to three R 4 .
  • X 1 is CH, n is 1, n1 is 1, and R 2 is (C 1 - 20 C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is N and X 3 is R 14 , n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is N and X 3 is R 14 , n is 1, n1 is 1, q is 0, 25 and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 13 and X 3 is N is R 14 , n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 13 and X 3 is N, n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 15 and X 3 is CR 14 , n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH, X 2 is CR 15 and X 3 is 30 CR14, n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R4.
  • X 1 is CH, X 2 is CR 13 and X 3 is CR 16 , n is 1, n1 is 1, q is 0, and R 2 is (C 1 -C 6 )alkyl substituted with one to three R 4 .
  • X 1 is CH
  • X 2 is CR 13 and X 3 is CR 16
  • n is 1
  • q is 0, and R 2 is (C 1 - C 6 )alkyl substituted with one to three R 4 . 5
  • Embodiment 1 A compound of Formula (I), wherein:
  • X1 is CR3
  • X 2 is N and X 3 is CR 14 ; or X 2 is CR 13 and X 3 is N; or X 2 is CR 15 and X 3 is CR 14 ; or X 2 is CR 13 and X 3 is 5 CR 16 ;
  • each R 1 is independently (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )hydroxyalkyl, CN, or halogen, or two R 1 together with the carbon atoms to which they are attached form (C 3 -C 7 )cycloalkyl or a 4- to 6- membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, or two R1, when on adjacent atoms, together with the atoms to which they are attached form a (C6-C10)aryl 10 ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S;
  • R 2 is (C 1 -C 6 )alkyl, (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 3 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the alkyl is optionally substituted with one or more R 4 ; and the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one or 15 more R 5 , or
  • R 1 and R 2 when on adjacent atoms, together with the atoms to which they are attached form a 5- or 6- membered heterocycloalkyl ring;
  • R 3 is H or R 3 is absent when is a double bond
  • each R 4 is independently selected from -C(O)OR 6 , -C(O)NR 6 R 6' , -NR 6 C(O)R 6' , halogen, -OH, -NH 2 , CN, 20 (C 6 -C 10 )aryl, 5- or 6-membered heteroaryl comprising 1 to 4 heteroatoms selected from O, N, and S, (C 3 -C 8 )cycloalkyl, and 4- to 7-membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl groups are optionally substituted with one or more R 7 ;
  • each R 5 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, 25 (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -NH 2 , CN,
  • R 5 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl 30 ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 , or
  • R 6 and R 6' are each independently H, (C 1 -C 6 )alkyl, or (C 6 -C 10 )aryl;
  • each R 7 is independently selected from (C 1 -C 6 )alkyl, (C 2 -C 6 )alkenyl, (C 2 -C 6 )alkynyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, -C(O)R 8 , -(CH 2 ) 0-3 C(O)OR 8 , -C(O)NR 8 R 9 , -NR 8 C(O)R 9 , - NR 8 C(O)OR 9 , -S(O) p NR 8 R 9 , -S(O) p R 12 , (C 1 -C 6 )hydroxyalkyl, halogen, -OH, -O(CH 2 ) 1-3 CN, -NH 2 , CN, -O(CH 2 )0-3(C6-C10)aryl, adamantyl,
  • R 7 when on adjacent atoms, together with the atoms to which they are attached form a (C 6 -C 10 )aryl ring or a 5- or 6-membered heteroaryl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 , or
  • R 7 together with the atoms to which they are attached form a (C 5 -C 7 ) cycloalkyl ring or a 5- to 7- 15 membered heterocycloalkyl ring comprising 1 to 3 heteroatoms selected from O, N, and S, optionally substituted with one or more R 10 ;
  • R 8 and R 9 are each independently H or (C 1 -C 6 )alkyl
  • each R 10 is independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl,
  • each R 11 is independently selected from CN, (C 1 -C 6 )alkoxy, (C 6 -C 10 )aryl, and 5- to 7-membered
  • heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S, wherein the aryl and heterocycloalkyl are optionally substituted with one or more substituents each independently selected from (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )hydroxyalkyl, 25 halogen, -OH, -NH 2 , and CN;
  • R 12 is (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 6 -C 10 )aryl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S;
  • R 13 is H, halogen, -OH, or -NH 2 ;
  • R 14 is H, (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, 30 halogen, -OH, -NH2, -NO2, or CN;
  • R 15 is halogen, -OH, or -NH 2 ;
  • R 16 is (C 1 -C 3 )alkyl, (C 1 -C 3 )alkoxy, (C 1 -C 3 )haloalkyl, (C 1 -C 3 )haloalkoxy, (C 1 -C 3 )hydroxyalkyl, halogen, -OH, -NH 2 , -NO 2 , or CN;
  • R x is H or D
  • 35 p is 0, 1, or 2;
  • n 0, 1, or 2;
  • n1 is 1 or 2, wherein n + n1 £ 3; and q is 0, 1, 2, 3, or 4;
  • Embodiment 2 The compound according to Embodiment 1, wherein R x is H.
  • Embodiment 3 The compound according to Embodiment 1 or 2, wherein X 2 is N and X 3 is CR 14 .
  • Embodiment 4 The compound according to Embodiment 1 or 2, wherein X 2 is CR 13 and X 3 is N.
  • Embodiment 5 The compound according to Embodiment 1 or 2, wherein X 2 is CR 15 , and X 3 is CR 14 .
  • Embodiment 6 The compound according to Embodiment 1 or 2, wherein X 2 is CR 13 , and X 3 is 10 CR16.
  • Embodiment 7 The compound according to Embodiment 1, having a Formula (Ia), Formula (Ib), Formula (Ic), or Formula (Id), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 8 The compound according to any one of Embodiments 1-7, wherein s a 15 double bond, X 1 is CR 3 , and R 3 is absent.
  • Embodiment 9 The compound according to any one of Embodiments 1-7, wherein a single bond, X 1 is CR 3 , and R 3 is H.
  • Embodiment 10 The compound according to Embodiment 1, having a Formula (Ie), Formula (If), Formula (Ig), or Formula (Ih), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, 20 stereoisomer, or tautomer thereof.
  • Embodiment 11 The compound according to any one of Embodiments 1-10, wherein n is 0, 1, or 2.
  • Embodiment 12 The compound according to any one of Embodiments 1-11, wherein n is 1 or 2.
  • Embodiment 13 The compound according to any one of Embodiments 1-12, wherein n is 1. 25
  • Embodiment 14 The compound according to Embodiment 1 having a Formula (Ii), Formula (Ij), Formula (Ik), or Formula (Il), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 15 The compound according to any one of Embodiments 1-14, wherein R 2 is (C 6 - C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected 30 from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • R 2 is (C 6 - C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected 30 from O, N, and S, wherein the aryl, heteroaryl, cycloalkyl, and heterocycloalkyl are optionally substituted with one to three R 5 .
  • Embodiment 16 The compound according to any one of Embodiments 1-14, wherein R 2 is (C 6 - C 10 )aryl, (C 3 -C 8 )cycloalkyl, or 5- to 7-membered heterocycloalkyl comprising 1 to 3 heteroatoms selected from O, N, and S.
  • Embodiment 17 The compound according to any one of Embodiments 1-14, wherein R 2 is (C 1 - C 6 )alkyl optionally substituted with one to three R 4 .
  • Embodiment 18 The compound according to any one of Embodiments 1-14, wherein R 2 is (C 1 - C 6 )alkyl substituted with one to three R 4 .
  • Embodiment 19 The compound according to any one of Embodiments 1-18, wherein q is 0, 1, or 2.
  • Embodiment 20 The compound according to any one of Embodiments 1-19, wherein q is 0 or 1.
  • Embodiment 21 The compound according to any one of Embodiments 1-20, wherein q is 0.
  • Embodiment 22 A compound selected from:
  • Embodiment 23 A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, 35 solvate, prodrug, stereoisomer, or tautomer thereof, and a pharmaceutically acceptable carrier or
  • Embodiment 24 The pharmaceutical composition according to Embodiment 23 further comprising at least one additional pharmaceutical agent.
  • Embodiment 25 The pharmaceutical composition according to Embodiment 23 or Embodiment 24 for use in the treatment of a disease or disorder that is affected by the reduction of IKZF2 protein 5 levels.
  • Embodiment 26 A method of degrading IKZF2 comprising administering to the patient in need thereof a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 27 A method of treating a disease or disorder that is affected by the modulation of 10 IKZF2 protein levels comprising administering to the patient in need thereof a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 28 A method of modulating IKZF2 protein levels comprising administering to the patient in need thereof a compound according to any one of Embodiments 1-22, or a pharmaceutically 15 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 29 A method of reducing the proliferation of a cell the method comprising, contacting the cell with a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and reducing IKZF2 protein levels.
  • Embodiment 30 A method of treating cancer comprising administering to the patient in need thereof a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Embodiment 31 The method according to Embodiment 30, wherein the cancer is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal 25 cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and gastrointestinal stromal tumor (GIST).
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • NPC nasopharyngeal 25 cancer
  • mssCRC microsatellite stable colorectal cancer
  • thymoma carcinoid
  • acute myelogenous leukemia and gastrointestinal stromal tumor (GIST).
  • GIST gastrointestinal stromal tumor
  • Embodiment 32 The method according to Embodiment 30, wherein the cancer is a cancer for which the immune response is deficient or an immunogenic cancer.
  • Embodiment 33 A method for reducing IKZF2 protein levels in a subject comprising the step of 30 administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt.
  • Embodiment 34 The method according to any one of Embodiments 26-33, wherein administering is performed orally, parentally, subcutaneously, by injection, or by infusion.
  • Embodiment 35 A compound according to any one of Embodiments 1-22, or a pharmaceutically 35 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of a disease or disorder that is affected by the reduction of IKZF2 protein levels.
  • Embodiment 36 Use of a compound according to any one of claims 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating a disease or disorder that is affected by the reduction of IKZF2 protein levels.
  • Embodiment 37 A compound according to any one of Embodiments 1-22, or a pharmaceutically 5 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating a disease or disorder associated with the reduction of IKZF2 protein levels.
  • Embodiment 38 Use of a compound according to any one of Embodiments 1-22, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of a disease or disorder associated with the reduction of IKZF2 protein levels.
  • Embodiment 39 The compound according to Embodiment 35 or 37 or the use according to
  • Embodiment 36 or 38 wherein the disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • NPC nasopharyngeal cancer
  • mssCRC microsatellite stable colorectal cancer
  • thymoma carcinoid
  • acute myelogenous leukemia and
  • GIST gastrointestinal stromal tumor
  • Embodiment 40 A compound, or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, selected from:
  • the compounds of the present disclosure are enantiomers. In some embodiments the compounds are the (S)-enantiomer. In other embodiments the compounds are the (R)-enantiomer. In yet other embodiments, the compounds of the present disclosure may be (+) or (-) 5 enantiomers.
  • the substituent may be in the E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans configuration. All tautomeric forms are also intended to be included.
  • the compounds of the disclosure may contain asymmetric or chiral centers and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the 15 disclosure as well as mixtures thereof, including racemic mixtures, form part of the present disclosure.
  • the present disclosure embraces all geometric and positional isomers. For example, if a compound of the disclosure incorporates a double bond or a fused ring, both the cis- and trans-forms, as well as mixtures, are embraced within the scope of the disclosure.
  • Each compound herein disclosed includes all the enantiomers that conform to the general structure of the compound.
  • the compounds may be in a racemic or enantiomerically pure form, or any other form in terms of stereochemistry.
  • the assay results may reflect the data collected for the racemic form, the enantiomerically pure form, or any other form in terms of 5 stereochemistry.
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active 10 compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
  • an appropriate optically active 10 compound e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride
  • converting e.g., hydrolyzing
  • some of the compounds of the disclosure may be atropisomers (e.g., substituted biaryls) and are considered as part of this disclosure.
  • Enantiomers
  • All stereoisomers (for example, geometric isomers, optical isomers, and the like) of the present compounds including those of the salts, solvates, esters, and prodrugs of the compounds as well as the 20 salts, solvates and esters of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this disclosure, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl).
  • each asymmetric atom has at least 50% enantiomeric excess, at least 60% enantiomeric excess, at least 70% enantiomeric excess, at least 80% enantiomeric excess, at least 90% enantiomeric excess, at least 95% enantiomeric excess, or at least 99% enantiomeric excess in the (R)- or (S)- configuration.
  • Substituents at atoms with unsaturated double bonds may, if possible, be present in cis-(Z)- or trans-(E)- form.
  • the compounds and intermediates may be isolated and used as the compound per se. Any formula 5 given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F, 31 P, 32 P, respectively.
  • the disclosure includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 13 C, and 14 C, are present.
  • isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of 15 patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F, 11 C or labeled compound may be particularly desirable for PET or SPECT studies.
  • substitution with heavier isotopes may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, reduced dosage requirements, reduced CYP450 inhibition (competitive or time dependent) or an 20 improvement in therapeutic index.
  • substitution with deuterium may modulate undesirable side effects of the undeuterated compound, such as competitive CYP450 inhibition, time dependent CYP450 inactivation, etc.
  • deuterium in this context is regarded as a substituent in compounds of the present disclosure.
  • the concentration of such a heavier isotope, specifically deuterium may be defined by the isotopic enrichment factor.
  • isotopic enrichment factor as used herein 25 means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this disclosure is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium 30 incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • Isotopically-labeled compounds of the present disclosure can generally be prepared by conventional techniques known to those skilled in the art or by carrying out the procedures disclosed in the 35 schemes or in the examples and preparations described below using an appropriate isotopically-labeled reagent in place of the non-isotopically labeled reagent.
  • Pharmaceutically acceptable solvates in accordance with the disclosure include those wherein the solvent of crystallization may be isotopically substituted, e.g., D 2 O, d 6 -acetone, d 6 -DMSO.
  • the present disclosure relates to compounds which are modulators of IKZF2 protein levels.
  • the compounds of the present disclosure decrease IKZF2 protein levels.
  • the compounds of the present disclosure reduce IKZF2 protein levels.
  • the compounds of the present disclosure are degraders of IKZF2.
  • the present disclosure relates to compounds, which are modulators of IKZF2 and IKZF4 protein levels.
  • the compounds of the present disclosure decrease IKZF2 and IKZF4 protein levels.
  • the compounds of the present disclosure reduce IKZF2 and IKZF4 protein 10 levels.
  • the compounds of the present disclosure are degraders of IKZF2.
  • the compounds of the disclosure are selective over other proteins.
  • “selective modulator”,“selective degrader”, or“selective compound” means, for example, a compound of the disclosure, that effectively modulates, decreases, or reduces the levels of a specific protein or degrades a specific protein to a greater extent than any other protein.
  • A“selective modulator”,“selective 15 degrader”, or“selective compound” can be identified, for example, by comparing the ability of a compound to modulate, decrease, or reduce the levels of or to degrade a specific protein to its ability to modulate, decrease, or reduce the levels of or to degrade other proteins.
  • the selectivity can be identified by measuring the AC 50 , EC 50 , or IC 50 of the compounds.
  • the compounds of the present application are selective IKZF2 modulators.
  • selective IKZF2 modulator “selective IKZF2 degrader”, or“selective IKZF2 compound” refers to a compound of the application, for example, that effectively modulates, decrease, or reduces the levels of IKZF2 protein or degrades IKZF2 protein to a greater extent than any other protein, particularly any protein (transcription factor) from the Ikaros protein family (e.g., IKZF1, IKZF3, IKZF4, and IKZF5).
  • IKZF1, IKZF3, IKZF4, and IKZF5 any protein (transcription factor) from the Ikaros protein family
  • A“selective IKZF2 modulator”,“selective IKZF2 degrader”, or“selective IKZF2 compound” can 25 be identified, for example, by comparing the ability of a compound to modulate IKZF2 protein levels to its ability to modulate levels of other members of the Ikaros protein family or other proteins. For example, a substance may be assayed for its ability to modulate IKZF2 protein levels, as well as IKZF1, IKZF3, IKZF4, IKZF5, and other proteins.
  • the selectivity can be identified by measuring the EC 50 of the compounds.
  • the selectivity can be identified by measuring the AC 50 of the 30 compounds.
  • a selective IKZF2 degrader is identified by comparing the ability of a compound to degrade IKZF2 to its ability to degrade other members of the Ikaros protein family or other proteins.
  • the compounds of the application are IKZF2 degraders that exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over 35 other proteins (e.g., IKZF1, IKZF3, IKZF4, and IKZF5). In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over other proteins.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over the other members of the Ikaros protein family (e.g., IKZF1, IKZF3, IKZF4, and IKZF5). In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over the other members 5 of the Ikaros protein family (e.g., IKZF1, IKZF3, IKZF4, and IKZF5).
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over IKZF1. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over IKZF1.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over IKZF3. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over IKZF3.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- 15 fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over IKZF4. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over IKZF4.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 over IKZF5. In various 20 embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 over IKZF5.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 and IKZF4 over the other members of the Ikaros protein family (e.g., IKZF1, IKZF3, and IKZF5).
  • the 25 compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 and IKZF4 over the other members of the Ikaros protein family (e.g., IKZF1, IKZF3, and IKZF5).
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 and IKZF4 over IKZF1. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the 30 degradation of IKZF2 and IKZF4 over IKZF1.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 and IKZF4 over IKZF3. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 and IKZF4 over IKZF3.
  • the compounds of the application exhibit at least 2-fold, 3-fold, 5-fold, 10- fold, 25-fold, 50-fold or 100-fold selectivity for the degradation of IKZF2 and IKZF4 over IKZF5. In various embodiments, the compounds of the application exhibit up to 1000-fold selectivity for the degradation of IKZF2 and IKZF4 over IKZF5.
  • the degradation of IKZF2 is measured by AC50.
  • Potency of can be determined by AC 50 value.
  • a compound with a lower AC 50 value, as determined 5 under substantially similar degradation conditions, is a more potent degrader relative to a compound with a higher AC 50 value.
  • the substantially similar conditions comprise determining degradation of protein levels in cells expressing the specific protein, or a fragment of any thereof.
  • the disclosure is directed to compounds as described herein and pharmaceutically acceptable salts, hydrates, solvates, prodrugs, stereoisomers, or tautomers thereof, and pharmaceutical compositions 10 comprising one or more compounds as described herein, or pharmaceutically acceptable salts, hydrates, solvates, prodrugs, stereoisomers, or tautomers thereof.
  • the compounds of the present disclosure may be made by a variety of methods, including standard chemistry. Suitable synthetic routes are depicted in the Schemes given below.
  • the compounds of the present disclosure may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes. In the schemes described below, it is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles or chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T.W. Greene and P.G.M. Wuts,“Protective Groups in Organic Synthesis”, 20 Third edition, Wiley, New York 1999). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection processes, as well as the reaction conditions and order of their execution, shall be consistent with the preparation of Compounds of Formula (I).
  • the present disclosure includes both possible stereoisomers (unless specified in the synthesis) and includes not only racemic compounds but the individual enantiomers and/or diastereomers as well.
  • a compound When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be affected by any suitable method known in 30 the art. See, for example,“Stereochemistry of Organic Compounds” by E.L. Eliel, S.H. Wilen, and L.N.
  • the compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes.
  • the compounds of the present disclosure can be prepared in a number of ways well known to those skilled in the art of organic synthesis.
  • compounds of the present disclosure can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art.
  • Preferred methods include but are not limited to those methods described below.
  • R1, R4, R16, Rx, X2, n, n1, and q are as defined in Formula (I).
  • Reaction of 1- 15 b and 1-c in the presence of a reducing agent e.g., sodium triacetoxyborohydride (NaB(OAc) 3 H), sodium cyanoborohydride (NaBH3CN), etc.
  • a reducing agent e.g., sodium triacetoxyborohydride (NaB(OAc) 3 H), sodium cyanoborohydride (NaBH3CN), etc.
  • a solvent e.g., DMF
  • Coupling of 1-d with iodide, bromide or tosylate 1-e using a catalyst e.g., NiBr 2 • (DME)
  • a catalyst e.g., NiBr 2 • (DME)
  • ligand picolinamide hydrochloride salt, 4,4'- di-tert-butyl-2,2'-dipyridyl, pyridine-2,6-bis(carboximidamide) dihydrochloride, 4- methoxypicolinimidamide hydrochloride,etc.
  • potassium iodide (KI) and manganese or zinc powder in a 20 solvent e.g., dimethylacetamide (DMA)
  • DMA dimethylacetamide
  • Removal of the amine protecting group (e.g., tert-butyloxycarbonyl (Boc)) on intermediate 1-f can be accomplished using a strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl) in a solvent (e.g., tetrahydrofuran (THF), 1,2-dichloroethane, dioxane or dichloromethane (DCM)) optionally at elevated temperature to provide I-g.
  • a strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl)
  • a solvent e.g., tetrahydrofuran (THF), 1,2-dichloroethane, dioxane or dichloromethane (DCM)
  • THF tetrahydrofuran
  • DCM dichloromethane
  • R 1 , R 4 , R x , X 2 , X 3 , n, n1, and q are as defined in Formula (I).
  • Coupling 15 of 2-c with iodide, bromide or tosylate 1-e using a catalyst e.g., NiBr 2 •(DME)
  • ligand e.g., picolinamide hydrochloride salt, 4,4'-di-tert-butyl-2,2'-dipyridyl, pyridine-2,6-bis(carboximidamide) dihydrochloride, 4- methoxypicolinimidamide hydrochloride,etc.
  • potassium iodide (KI) and manganese or zinc powder in a solvent e.g., dimethylacetamide (DMA) optionally at elevated temperature provides 2-d.
  • a solvent e.g., dimethylacetamide (DMA)
  • Removal of the amine protecting group (e.g., tert-butyloxycarbonyl (Boc)) on intermediate 2-d can be accomplished using 20 a strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl) in a solvent (e.g., tetrahydrofuran (THF), 1,2,-dichloroethane, dioxane or dichloromethane (DCM)) optionally at elevated temperature to provide 2-e.
  • a strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl)
  • a solvent e.g., tetrahydrofuran (THF), 1,2,-dichloroethane, dioxane or dichloromethane (DCM)
  • THF tetrahydrofuran
  • DCM dichloromethane
  • Reductive amination of 2-e with aldehyde or ketone 2-f provides a compound of Formula (I
  • R 1 , R 4 , R x , X 1 , X 2 , X 3 , n, n1, and q are as defined in Formula (I).
  • Removal of the amine protecting group (e.g., tert-butyloxycarbonyl (Boc)) on intermediate 3-b can be accomplished using a 10 strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl) in a solvent (e.g., tetrahydrofuran (THF), 1,2,-dichloroethane, dioxane or dichloromethane (DCM)) optionally at elevated temperature to provide 3-c.
  • a 10 strong acid such as trifluoroacetic acid (TFA) or hydrochloric acid (HCl)
  • a solvent e.g., tetrahydrofuran (THF), 1,2,-dichloroethane, dioxane or dichloromethane (DCM)
  • THF tetrahydrofuran
  • DCM dichloromethane
  • Compounds of Formula (I) where is a double bond, X 1 is CR 3 , R 3 is absent, and R 2 is a substituted alkyl can be 15 obtained by alkylation of 3-c with an alkyl halide 3-d in the presence of a base (e.g., NEt 3 , Cs 2 CO 3 , etc.), in a solvent (e.g., DCM, DMF, etc.), and optionally at elevated temperature.
  • a base e.g., NEt 3 , Cs 2 CO 3 , etc.
  • solvent e.g., DCM, DMF, etc.
  • a mixture of enantiomers, diastereomers, and cis/trans isomers resulting from the process described above can be separated into their single components by chiral salt technique, chromatography using normal phase, reverse phase or chiral column, depending on the nature of the separation.
  • Any resulting racemates of compounds of the present disclosure or of intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound.
  • a basic moiety may thus be employed to resolve the compounds of the present disclosure into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., 25 tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-O,O'-p-toluoyl tartaric acid, mandelic acid, malic acid, or camphor-10-sulfonic acid.
  • Racemic compounds of the present disclosure or racemic intermediates can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high pressure liquid chromatography
  • Any resulting mixtures of stereoisomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, 5 diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
  • Another aspect of the disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder in a patient associated with or affected by modulation of IKZF2 protein levels.
  • the method comprises administering to a patient in need of a treatment for diseases or disorders 15 associated with modulation of IKZF2 protein levels an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the disclosure relates to a method of treating, preventing, inhibiting, or 20 eliminating a disease or disorder that is affected by the reduction of or decrease in IKZF2 protein levels.
  • the method comprises administering to a patient in need of a treatment for diseases or disorders affected by the reduction of IKZF2 protein levels an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a Compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 25 prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment, 30 prevention, inhibition or elimination of a disease or disorder that is associated with or affected by the modulation of IKZF2 protein levels.
  • the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 35 prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment, prevention, inhibition or elimination of a disease or disorder that is affected by the reduction of or a decrease in IKZF2 protein levels.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating, preventing, inhibiting, or 5 eliminating a disease or disorder that is associated with or affected by the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • the present disclosure is directed to a method of modulating, reducing, or decreasing IKZF2 protein levels.
  • the method involves administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 10 prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF2 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 protein.
  • IKZF2 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 protein mediated by an E3 ligase.
  • Another aspect of the present disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder in a patient associated with the reduction of or decrease in IKZF2 protein levels, the method comprising administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, 20 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure also relates to the use of a degrader of IKZF2 for the preparation of a medicament used in the treatment, prevention, inhibition or elimination of a IKZF2-dependent disease or disorder, wherein the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a Compound 25 of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a method for treating, preventing, inhibiting, or eliminating a IKZF2-dependent disease or disorder, wherein the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer 30 thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a method for the manufacture of a medicament for treating, preventing, inhibiting, or eliminating a IKZF2-dependent disease or disorder mediated, wherein the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, 35 solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating a disease 5 or disorder associated with the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • IKZF2 levels are modulated through degradation of the IKZF2 protein.
  • IKZF2 protein levels are modulated through degradation of the IKZF2 protein mediated by an E3 ligase.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a 10 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating a disease associated with the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • IKZF2 levels are modulated, reduced, or decreased through degradation of the IKZF2 protein.
  • IKZF2 protein 15 levels are modulated, reduced, or decreased through degradation of the IKZF2 protein mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 20 prodrug, stereoisomer, or tautomer thereof, in the treatment of a disease associated with the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • IKZF2 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 protein.
  • IKZF2 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 protein mediated by an E3 ligase.
  • the present disclosure relates to a method of inhibiting IKZF2 activity through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 30 prodrug, stereoisomer, or tautomer thereof, for inhibiting IKZF2 activity through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, 35 or tautomer thereof, for use in the inhibition of IKZF2 activity through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for inhibiting IKZF2 activity through 5 degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a method of inhibiting IKZF2 and IKZF4 activity through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a 10 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for inhibiting IKZF2 and IKZF4 activity through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the inhibition of IKZF2 and IKZF4 activity through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for inhibiting IKZF2 and IKZF4 activity through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation 25 is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder associated with the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, 30 stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure is directed to a method of modulating, reducing, or decreasing IKZF2 and IKZF4 protein levels.
  • the method involves administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 35 prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are modulated through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder associated with modulation of, reduction of, or a decrease in IKZF4 protein 5 levels.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 proteins.
  • 10 IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 protein mediated by an E3 ligase.
  • the present disclosure is directed to a method of modulating, reducing, or decreasing IKZF4 protein levels.
  • the method involves administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 15 prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 proteins.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 protein mediated by an E3 ligase.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for treating, preventing, inhibiting, or eliminating a disease or disorder associated with modulation of, reduction of, or a decrease in IKZF4 protein levels.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 proteins.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 protein mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a 30 compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating, preventing, inhibiting, or eliminating a disease or disorder associated with modulation of, reduction of, or a decrease in IKZF4 protein levels.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 proteins.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 35 protein mediated by an E3 ligase.
  • the present disclosure is directed to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating, preventing, inhibiting, or eliminating a disease or disorder associated with modulation of, reduction of, or a decrease in IKZF4 protein levels.
  • IKZF4 protein levels are modulated, reduced, 5 or decreased through degradation of the IKZF4 proteins.
  • IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF4 protein mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder associated with a decrease in IKZF2 and IKZF4 protein levels.
  • the method comprises administering to a patient in need of a treatment for diseases or disorders associated with a 10 decrease of IKZF2 and IKZF4 protein levels an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure also relates to the use of a modulator of IKZF2 and IKZF4 protein levels for 15 the preparation of a medicament used in the treatment, prevention, inhibition or elimination of a IKZF2 and IKZF4-dependent disease or disorder, wherein the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a method for20 the manufacture of a medicament for treating, preventing, inhibiting, or eliminating a IKZF2 and IKZF4- dependent disease or disorder, wherein the medicament comprises a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating a disease associated with the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically 35 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating a disease associated with the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of a disease associated with the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • IKZF2 and 10 IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are modulated, reduced, or decreased through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 15 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of an IKZF2-dependent disease or disorder by reducing or decreasing IKZF2 protein levels, wherein reduction or decrease of IKZF2 protein levels treats the IKZF2- dependent disease or disorder.
  • the present disclosure the use of a compound of Formula (I), or a 20 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of an IKZF2-dependent disease or disorder by reducing or decreasing IKZF2 protein levels wherein reduction of or decrease in IKZF2 protein levels treats the IKZF2-dependent disease or disorder.
  • the present disclosure the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating an IKZF2- dependent disease or disorder by reducing or decreasing IKZF2 protein levels wherein reduction of or 30 decrease in IKZF2 protein levels treats the IKZF2-dependent disease or disorder.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of an IKZF2 and IKZF4-dependent disease or disorder by 35 reducing or decreasing IKZF2 and IKZF4 protein levels wherein the reduction of or decrease in IKZF2 and IKZF4 protein levels treats the IKZF2 and IKZF4-dependent disease or disorder.
  • the present disclosure the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of an IKZF2 and IKZF4-dependent disease or 5 disorder by reducing or decreasing IKZF2 and IKZF4 protein levels wherein the reduction of or decrease in IKZF2 and IKZF4 protein levels treats the IKZF2 and IKZF4-dependent disease or disorder.
  • the present disclosure the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 10 prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating an IKZF2 and IKZF4-dependent disease or disorder by reducing or decreasing IKZF2 and IKZF4 protein levels wherein the reduction of or decrease in IKZF2 and IKZF4 protein levels treats the IKZF2 and IKZF4-dependent disease or disorder.
  • Another aspect of the disclosure relates to a method of treating cancer.
  • the method comprises 15 administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to the use of a compound of Formula (I), or a 20 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of treating cancer.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 25 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating cancer.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, 30 or tautomer thereof, for use in the treatment of cancer.
  • Another aspect of the disclosure relates to a method of treating an IKZF2-dependent cancer.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 35 prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of treating an IKZF2-dependent cancer.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 5 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent cancer.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, 10 or tautomer thereof, for use in the treatment of an IKZF2-dependent cancer.
  • Another aspect of the disclosure relates to a method of treating an IKZF2-dependent and IKZF4- dependent cancer.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically 15 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of treating an IKZF2-dependent and IKZF4- 20 dependent cancer.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent and 25 IKZF4-dependent cancer.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of an IKZF2-dependent and IKZF4-dependent cancer.
  • Another aspect of the disclosure relates to a method of treating a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer 35 thereof.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of treating a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 protein levels
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 5 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically 10 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 protein levels.
  • Another aspect of the disclosure relates to a method of treating a cancer affected by the modulation 15 of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the treatment of treating a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of a cancer affected by the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a 5 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for degrading IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 10 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the degradation IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically 15 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for degrading IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a method of modulating IKZF2 protein levels 20 through degradation of IKZF2.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for modulating IKZF2 protein levels through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the modulation IKZF2 protein levels through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for modulating IKZF2 protein levels through degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of treating an IKZF2-dependent disease or 5 disorder in a patient in need thereof by modulating IKZF2 protein levels through the degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 10 prodrug, stereoisomer, or tautomer thereof, for treating an IKZF2-dependent disease or disorder in a patient in need thereof by modulating IKZF2 protein levels through the degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 15 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating an IKZF2-dependent disease or disorder in a patient in need thereof, by modulating IKZF2 protein levels through the degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically 20 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent disease or disorder in a patient in need thereof by modulating IKZF2 protein levels through the degradation of IKZF2.
  • IKZF2 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of reducing the proliferation of a cell, the method comprising contacting the cell with a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, that reduces IKZF2 protein levels.
  • IKZF2 protein levels are reduced through 30 degradation of the IKZF2 protein.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein mediated by an E3 ligase.
  • the present disclosure relates to the use a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 35 prodrug, stereoisomer, or tautomer thereof, for reducing the proliferation of a cell by reducing IKZF2 protein levels.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 5 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in reducing the proliferation of a cell by IKZF 2 protein levels.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for reducing the proliferation of a cell by reducing IKZF2 protein levels.
  • IKZF2 protein levels are reduced through 15 degradation of the IKZF2 protein.
  • IKZF2 protein levels are reduced through degradation of the IKZF2 protein mediated by an E3 ligase.
  • the disclosure relates to a method of treating, preventing, inhibiting, or eliminating a disease or disorder that is affected by the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the method comprises administering to a patient in need thereof an 20 effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a 25 composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment, prevention, inhibition or elimination of a disease or disorder that is affected by the modulation of IKZF2 and IKZF4 protein levels.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 30 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating, preventing, inhibiting, or eliminating a disease or disorder that is affected by the modulation of, the reduction of, or a decrease in IKZF2 and IKZF4 protein levels.
  • the disclosure relates to the use a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for the treatment, prevention, inhibition or elimination of a disease or disorder that is affected by the reduction of or a decrease in IKZF2 and IKZF4 protein levels.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 5 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating, preventing, inhibiting, or eliminating a disease or disorder that is affected by the reduction of or a decrease in IKZF2 and IKZF4 protein levels.
  • Another aspect of the disclosure relates to a method of degrading IKZF2 and IKZF4.
  • the method comprises administering to a patient in need thereof an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • IKZF2 and IKZF4 protein degradation 15 is mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for degrading IKZF2 and IKZF4.
  • IKZF2 20 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the degradation IKZF2 and IKZF4.
  • IKZF2 and IKZF4 25 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for degrading IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a method of modulating IKZF2 and IKZF4 protein levels through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to the use of a compound of Formula (I), or a 35 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for modulating IKZF2 and IKZF4 protein levels through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 5 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the modulation of IKZF2 and IKZF4 protein levels through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 10 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for modulating IKZF2 and IKZF4 protein levels through degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of treating an IKZF2-dependent and IKZF4- dependent disease or disorder in a patient in need thereof by modulating IKZF2 and IKZF4 protein levels through the degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to the use of a compound of Formula (I), or a 20 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for treating an IKZF2-dependent and IKZF4-dependent disease or disorder in a patient in need thereof by modulating IKZF2 and IKZF4 protein levels through the degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is 25 mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in treating an IKZF2-dependent and IKZF4-dependent disease or disorder in a 30 patient in need thereof by modulating IKZF2 and IKZF4 protein levels through the degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer,35 or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent or IKZF4- dependent disease or disorder in a patient in need thereof by modulating IKZF2 and IKZF4 protein levels through the degradation of IKZF2 and IKZF4.
  • IKZF2 and IKZF4 protein degradation is mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a method of reducing the proliferation of a cell, the method comprising contacting the cell with a compound of Formula (I), or a pharmaceutically acceptable 5 salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, and reducing IKZF2 and IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins mediated by 10 an E3 ligase.
  • the present disclosure relates to the use a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for reducing the proliferation of a cell by reducing IKZF2 and 15 IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 20 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in reducing the proliferation of a cell by reducing IKZF2 and IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for reducing the proliferation of a cell by reducing IKZF2 and IKZF4 protein levels.
  • IKZF2 and IKZF4 protein levels are 30 reduced through degradation of the IKZF2 and IKZF4 proteins.
  • IKZF2 and IKZF4 protein levels are reduced through degradation of the IKZF2 and IKZF4 proteins mediated by an E3 ligase.
  • the present disclosure relates to a method for treating an IKZF2-dependent disease or disorder.
  • the method comprises the step of administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, 35 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of an IKZF2-dependent disease or disorder.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating an IKZF2-dependent disease or disorder.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising 10 a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent disease or disorder.
  • the present disclosure relates to a method for treating an IKZF2-dependent and IKZF4-dependent disease or disorder.
  • the method comprises the step of administering to a subject in need 15 thereof a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 20 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the treatment of an IKZF2-dependent and IKZF4-dependent disease or disorder.
  • the present disclosure relates to the use of a compound of Formula (I), or a 25 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating an IKZF2- dependent and IKZF4-dependent disease or disorder.
  • Another aspect of the disclosure relates to a compound of Formula (I), or a pharmaceutically 30 acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for use in the manufacture of a medicament for treating an IKZF2-dependent and IKZF4-dependent disease or disorder.
  • the present disclosure relates to a method of reducing IKZF2 protein levels.
  • the 35 method comprises administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the present disclosure relates to a method of reducing IKZF2 and IKZF4 protein levels.
  • the method comprises administering to the patient in need thereof a compound of Formula (I), or a 5 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a 10 compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in the reduction of IKZF2 protein levels.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 15 prodrug, stereoisomer, or tautomer thereof for use in the reduction of IKZF2 and IKZF4 protein levels.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition, in the manufacture of a medicament for reducing IKZF2 protein levels.
  • Another aspect of the present disclosure relates to the use of a compound of Formula (I), or a 20 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for reducing IKZF2 and IKZF4 protein levels.
  • the present disclosure relates to a method of reducing IKZF2 protein levels, 25 wherein reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • the method comprises administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the present disclosure relates to a method of reducing IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the method comprises administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, 35 prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in the reduction of IKZF2 protein levels, wherein reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a 5 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in the reduction of IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the present disclosure relates to the use of a compound of Formula (I), or a 10 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition, in the manufacture of a medicament for reducing IKZF2 protein levels, wherein reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • Another aspect of the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a 15 composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for reducing IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the present disclosure relates to a method of treating a disease or disorder by 20 reducing IKZF2 protein levels, wherein reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • the method comprises administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • Another aspect of the present disclosure relates to a method of treating a disease or disorder by reducing IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the method comprises administering to the patient in need thereof a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable 30 salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof.
  • the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in the treatment of a disease or disorder by reducing IKZF2 protein levels, wherein 35 reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • Another aspect of the present disclosure relates to a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in the treatment of a disease or disorder by reducing IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the present disclosure relates to the use of a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition, in the manufacture of a medicament for treating a disease or disorder by reducing IKZF2 protein levels, wherein reduction of IKZF2 protein levels treats or ameliorates the disease or disorder.
  • Another aspect of the present disclosure relates to the use of a compound of Formula (I), or a 10 pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof or a composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, in the manufacture of a medicament for treating a disease or disorder by reducing IKZF2 and IKZF4 protein levels, wherein reduction of IKZF2 and IKZF4 protein levels treats or ameliorates the disease or disorder.
  • the compounds of the present disclosure can be used for the treatment, of a disease or disorder selected from liposarcoma, neuroblastoma, glioblastoma, bladder cancer, adrenocortical cancer, multiple myeloma, colorectal cancer, non-small cell lung cancer, Human Papilloma Virus-associated cervical, oropharyngeal, penis, anal, thyroid, or vaginal cancer or Epstein-Barr Virus-associated nasopharyngeal carcinoma, gastric cancer, rectal cancer, thyroid cancer, Hodgkin lymphoma or diffuse large B-cell 20 lymphoma.
  • a disease or disorder selected from liposarcoma, neuroblastoma, glioblastoma, bladder cancer, adrenocortical cancer, multiple myeloma, colorectal cancer, non-small cell lung cancer, Human Papilloma Virus-associated cervical, oropharyngeal, penis, anal, thyroid, or
  • the cancer is selected from prostate cancer, breast carcinoma, lymphomas, leukaemia, myeloma, bladder carcinoma, colon cancer, cutaneous melanoma, hepatocellular carcinoma, endometrial cancer, ovarian cancer, cervical cancer, lung cancer, renal cancer, glioblastoma multiform, glioma, thyroid cancer, parathyroid tumor, nasopharyngeal cancer, tongue cancer, pancreatic cancer, esophageal cancer, cholangiocarcinoma, gastric cancer, soft tissue sarcomas, rhabdomyosarcoma (RMS), synovial sarcoma, 25 osteosarcoma, rhabdoid cancers, cancer for which the immune response is deficient, an immunogenic cancer, and Ewing’s sarcoma.
  • prostate cancer breast carcinoma, lymphomas, leukaemia, myeloma, bladder carcinoma, colon cancer, cutaneous melanoma, hepatocellular carcinoma, endometrial cancer
  • the IKZF2-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, and gastrointestinal stromal tumor (GIST).
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • NPC nasopharyngeal cancer
  • mssCRC microsatellite stable colorectal cancer
  • thymoma thymoma
  • carcinoid gastrointestinal stromal tumor
  • the cancer is selected from 30 non-small cell lung cancer (NSCLC), melanoma, triple-negative breast cancer (TNBC), nasopharyngeal cancer (NPC), microsatellite stable colorectal cancer (mssCRC), thymoma, carcinoid, acute myelogenous leukemia, and gastrointestinal stromal tumor (GIST).
  • the IKZF2-dependent disease or disorder is a disease or disorder is selected from non-small cell lung cancer (NSCLC), melanoma, triple- negative breast cancer (TNBC), nasopharyngeal cancer (NPC), and microsatellite stable colorectal cancer 35 (mssCRC).
  • the disclosed compounds of the disclosure can be administered in effective amounts to treat or prevent a disorder and/or prevent the development thereof in subjects.
  • G. Administration, Pharmaceutical Compositions, and Dosing of Compounds of the Disclosure Administration of the disclosed compounds can be accomplished via any mode of administration for therapeutic agents. These modes include systemic or local administration such as oral, nasal, parenteral, transdermal, subcutaneous, vaginal, buccal, rectal or topical administration modes.
  • compositions can be in solid, semi-solid or liquid dosage form, such as, for example, injectables, tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • injectables tablets, suppositories, pills, time-release capsules, elixirs, tinctures, emulsions, syrups, powders, liquids, suspensions, or the like, sometimes in unit dosages and consistent with conventional pharmaceutical practices.
  • they can also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous or intramuscular form, and all using 10 forms well known to those skilled in the pharmaceutical arts.
  • Illustrative pharmaceutical compositions are tablets and gelatin capsules comprising a compound of the disclosure and a pharmaceutically acceptable carrier, such as a) a diluent, e.g., purified water, triglyceride oils, such as hydrogenated or partially hydrogenated vegetable oil, or mixtures thereof, com oil, olive oil, sunflower oil, safflower oil, fish oils, such as EPA or DHA, or their esters or triglycerides or 15 mixtures thereof, omega-3 fatty acids or derivatives thereof, lactose, dextrose, sucrose, mannitol, sorbitol, cellulose, sodium, saccharin, glucose and/or glycine; b) a lubricant, e.g., silica, talcum, stearic acid, its magnesium or calcium salt, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and/or polyethylene glycol; for tablets also
  • Liquid, particularly injectable, compositions can, for example, be prepared by dissolution, dispersion, etc.
  • the disclosed compound is dissolved in or mixed with a pharmaceutically 30 acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form an injectable isotonic solution or suspension.
  • a pharmaceutically 30 acceptable solvent such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like.
  • Proteins such as albumin, chylomicron particles, or serum proteins can be used to solubilize the disclosed compounds.
  • the disclosed compounds can be also formulated as a suppository that can be prepared from fatty emulsions or suspensions; using polyalkylene glycols such as propylene glycol, as the carrier.
  • Liposome delivery systems such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, containing cholesterol, stearylamine or phosphatidylcholines.
  • a film of lipid components is hydrated with an aqueous solution of drug to a form lipid layer encapsulating the drug, as described in U.S. Pat. No. 5,262,564 which is hereby incorporated by reference in its entirety.
  • Disclosed compounds can also be delivered by the use of monoclonal antibodies as individual 5 carriers to which the disclosed compounds are coupled.
  • the disclosed compounds can also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspanamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the disclosed compounds can be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for 10 example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and cross-linked or amphipathic block copolymers of hydrogels.
  • a polymer e.g., a polycarboxylic acid polymer, or a polyacrylate.
  • Parental injectable administration is generally used for subcutaneous, intramuscular or intravenous 15 injections and infusions.
  • Injectables can be prepared in conventional forms, either as liquid solutions or suspensions or solid forms suitable for dissolving in liquid prior to injection.
  • compositions comprising a compound of Formula (I), and a pharmaceutically acceptable carrier.
  • the pharmaceutical acceptable carrier may further include an excipient, diluent, or surfactant.
  • compositions can be prepared according to conventional mixing, granulating or coating methods, respectively, and the present pharmaceutical compositions can contain from about 0.1% to about 99%, from about 5% to about 90%, or from about 1% to about 20% of the disclosed compound by weight or volume.
  • the disclosure provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of the present disclosure.
  • the 25 kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the disclosure may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the 30 separate compositions against one another.
  • the kit of the disclosure typically comprises directions for administration.
  • the dosage regimen utilizing the disclosed compound is selected in accordance with a variety of factors including type, species, age, weight, sex, and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal or hepatic function of the patient; and the 35 particular disclosed compound employed.
  • a physician or veterinarian of ordinary skill in the art can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Effective dosage amounts of the disclosed compounds, when used for the indicated effects range from about 0.5 mg to about 5000 mg of the disclosed compound as needed to treat the condition.
  • compositions for in vivo or in vitro use can contain about 0.5, 5, 20, 50, 75, 100, 150, 250, 500, 750, 1000, 1250, 2500, 3500, or 5000 mg of the disclosed compound, or, in a range of from one amount to another 5 amount in the list of doses.
  • the compositions are in the form of a tablet that can be scored.
  • the compounds of the disclosure can be administered in therapeutically effective amounts in a combinational therapy with one or more therapeutic agents (pharmaceutical combinations) or modalities, 10 e.g., non-drug therapies.
  • therapeutic agents pharmaceutical combinations
  • modalities e.g., non-drug therapies.
  • synergistic effects can occur with other cancer agents.
  • dosages of the co- administered compounds will of course vary depending on the type of co-drug employed, on the specific drug employed, on the condition being treated and so forth.
  • the compounds can be administered simultaneously (as a single preparation or separate 15 preparation), sequentially, separately, or over a period of time to the other drug therapy or treatment modality.
  • a combination therapy envisions administration of two or more drugs during a single cycle or course of therapy.
  • a therapeutic agent is, for example, a chemical compound, peptide, antibody, antibody fragment or nucleic acid, which is therapeutically active or enhances the therapeutic activity when administered to a patient in combination with a compound of the present disclosure.
  • a compound of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure can be combined with other therapeutic agents, such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • other therapeutic agents such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, 25 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof of the present disclosure are administered in combination with one or more second agent(s) selected from a PD-1 inhibitor, a PD-L1 inhibitor, a LAG- 3 inhibitor, a cytokine, an A2A antagonist, a GITR agonist, a TIM-3 inhibitor, a STING agonist, and a TLR7 agonist, to treat a disease, e.g., cancer.
  • a second agent(s) selected from a PD-1 inhibitor, a PD-L1 inhibitor, a LAG- 3 inhibitor, a cytokine, an A2A antagonist, a GITR agonist, a TIM-3 inhibitor, a STING agonist, and a TLR7 agonist
  • one or more chemotherapeutic agents are used in combination with the 30 compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for treating a disease, e.g., cancer
  • said chemotherapeutic agents include, but are not limited to, anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5- deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), 35 cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (C
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more other anti-HER 2 antibodies, e.g., trastuzumab, pertuzumab, margetuximab, 15 or HT-19 described above, or with other anti-HER 2 conjugates, e.g., ado-trastuzumab emtansine (also known as Kadcyla®, or T-DM1).
  • anti-HER 2 antibodies e.g., trastuzumab, pertuzumab, margetuximab, 15 or HT-19 described above
  • other anti-HER 2 conjugates e.g., ado-trastuzumab emtansine (also known as Kadcyla®, or T-DM1).
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more tyrosine kinase inhibitors, including but not limited to, EGFR inhibitors, 20 Her3 inhibitors, IGFR inhibitors, and Met inhibitors, for treating a disease, e.g., cancer.
  • tyrosine kinase inhibitors including but not limited to, EGFR inhibitors, 20 Her3 inhibitors, IGFR inhibitors, and Met inhibitors, for treating a disease, e.g., cancer.
  • tyrosine kinase inhibitors include but are not limited to, Erlotinib hydrochloride (Tarceva®); Linifanib (N-[4-(3-amino-1H-indazol-4-yl)phenyl]-N'-(2-fluoro-5-methylphenyl)urea, also known as ABT 869, available from Genentech); Sunitinib malate (Sutent®); Bosutinib (4-[(2,4-dichloro- 5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methylpiperazin-1-yl)propoxy]quinoline-3-carbonitrile, also 25 known as SKI-606, and described in US Patent No.
  • Tarceva® Erlotinib hydrochloride
  • Linifanib N-[4-(3-amino-1H-indazol-4-yl)phenyl]-N'-(2-fluoro-5-methylphenyl
  • Epidermal growth factor receptor (EGFR) inhibitors include but are not limited to, Erlotinib hydrochloride (Tarceva®), Gefitinib (Iressa®); N-[4-[(3-Chloro-4-fluorophenyl)amino]-7-[[(3''S'')- 30 tetrahydro-3-furanyl]oxy]-6-quinazolinyl]-4(dimethylamino)-2-butenamide, Tovok®); Vandetanib (Caprelsa®); Lapatinib (Tykerb®); (3R,4R)-4-Amino-1-((4-((3-methoxyphenyl)amino)pyrrolo[2,1- f][1,2,4]triazin-5-yl)methyl)piperidin-3-ol (BMS690514); Canertinib dihydrochloride (CI-1033); 6-[4-[(4- Ethyl-1-
  • EGFR antibodies include but are not limited to, Cetuximab (Erbitux®); Panitumumab (Vectibix®); Matuzumab (EMD-72000); Nimotuzumab (hR3); Zalutumumab; TheraCIM h-R3; MDX0447 (CAS 5 339151-96-1); and ch806 (mAb-806, CAS 946414-09-1).
  • HER 2 inhibitors include but are not limited to, Neratinib (HKI-272, (2E)-N-[4-[[3-chloro-4- [(pyridin-2-yl)methoxy]phenyl]amino]-3-cyano-7-ethoxyquinolin-6-yl]-4-(dimethylamino)but-2-enamide, and described PCT Publication No.
  • HER3 inhibitors include but are not limited to, LJM716, MM-121, AMG-888, RG7116, REGN- 1400, AV-203, MP-RM-1, MM-111, and MEHD-7945A.
  • MET inhibitors include but are not limited to, Cabozantinib (XL184, CAS 849217-68-1); Foretinib (GSK1363089, formerly XL880, CAS 849217-64-7); Tivantinib (ARQ197, CAS 1000873-98-2); 1-(2-20 Hydroxy-2-methylpropyl)-N-(5-(7-methoxyquinolin-4-yloxy)pyridin-2-yl)-5-methyl-3-oxo-2-phenyl-2,3- dihydro-1H-pyrazole-4-carboxamide (AMG 458); Cryzotinib (Xalkori®, PF-02341066); (3Z)-5-(2,3- Dihydro-1H-indol-1-ylsulfonyl)-3-( ⁇ 3,5-dimethyl-4-[(4-methylpiperazin-1-yl)carbonyl]-1H-pyrrol-2- yl
  • 35 IGFR inhibitors include but are not limited to, BMS-754807, XL-228, OSI-906, GSK0904529A, A-928605, AXL1717, KW-2450, MK0646, AMG479, IMCA12, MEDI-573, and BI836845. See e.g., Yee, JNCI, 104; 975 (2012) for review.
  • the compounds of Formula (I) of the present disclosure are used in combination with one or more proliferation signaling pathway inhibitors, including but not limited to, MEK inhibitors, BRAF inhibitors, PI3K/Akt inhibitors, SHP2 inhibitors, and also mTOR inhibitors, and CDK inhibitors, for treating a disease, e.g., cancer.
  • one or more proliferation signaling pathway inhibitors including but not limited to, MEK inhibitors, BRAF inhibitors, PI3K/Akt inhibitors, SHP2 inhibitors, and also mTOR inhibitors, and CDK inhibitors, for treating a disease, e.g., cancer.
  • mitogen-activated protein kinase (MEK) inhibitors include but are not limited to, XL- 518 (also known as GDC-0973, Cas No. 1029872-29-4, available from ACC Corp.); 2-[(2-Chloro-4- iodophenyl)amino]-N-(cyclopropylmethoxy)-3,4-difluoro-benzamide (also known as CI-1040 or PD184352 and described in PCT Publication No.
  • XL- 518 also known as GDC-0973, Cas No. 1029872-29-4, available from ACC Corp.
  • 2-[(2-Chloro-4- iodophenyl)amino]-N-(cyclopropylmethoxy)-3,4-difluoro-benzamide also known as CI-1040 or PD184352 and described in PCT Publication No.
  • N-[3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]-6-methoxyphenyl]-1-[(2R)-2,3-dihydroxypropyl]- cyclopropanesulfonamide also known as RDEA119 or BAY869766 and described in PCT Publication No.
  • BRAF inhibitors include, but are not limited to, Vemurafenib (or Zelboraf®), GDC-0879, PLX- 4720 (available from Symansis), Dabrafenib (or GSK2118436), LGX 818, CEP-32496, UI-152, RAF 265, Regorafenib (BAY 73-4506), CCT239065, or Sorafenib (or Sorafenib Tosylate, or Nexavar®), or Ipilimumab (or MDX-010, MDX-101, or Yervoy).
  • Phosphoinositide 3-kinase (PI3K) inhibitors include, but are not limited to, 4-[2-(1H-Indazol-4-yl)- 6-[[4-(methylsulfonyl)piperazin-1-yl]methyl]thieno[3,2-d]pyrimidin-4-yl]morpholine (also known as GDC0941, RG7321, GNE0941, Pictrelisib, or Pictilisib; and described in PCT Publication Nos.
  • mTOR inhibitors include but are not limited to, Temsirolimus (Torisel®); Ridaforolimus (formally known as deferolimus, (1R,2R,4S)-4-[(2R)-2 [(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28Z,30S,32S,35R)-1,18-dihydroxy-19,30-dimethoxy- 15,17,21,23, 29,35-hexamethyl-2,3,10,14,20-pentaoxo-11,36-dioxa-4-azatricyclo[30.3.1.04,9] 5 hexatriaconta-16,24,26,28-tetraen-12-yl]propyl]-2-methoxycyclohexyl dimethylphosphinate, also known as AP23573 and MK8669, and described in PCT Publication No.
  • CDK inhibitors include but are not limited to, Palbociclib (also known as PD-0332991, Ibrance®, 6-Acetyl-8-cyclopentyl-5-methyl-2- ⁇ [5-(1-piperazinyl)-2-pyridinyl]amino ⁇ pyrido[2,3-d]pyrimidin- 15 7(8H)-one).
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more pro-apoptotics, including but not limited to, IAP inhibitors, BCL2 inhibitors, MCL1 inhibitors, TRAIL agents, CHK inhibitors, for treating a disease, e.g., cancer.
  • pro-apoptotics including but not limited to, IAP inhibitors, BCL2 inhibitors, MCL1 inhibitors, TRAIL agents, CHK inhibitors, for treating a disease, e.g., cancer.
  • IAP inhibitors include but are not limited to, LCL161, GDC-0917, AEG-35156, AT406, and TL32711.
  • Other examples of IAP inhibitors include but are not limited to those disclosed in WO04/005284, WO 04/007529, WO05/097791, WO 05/069894, WO 05/069888, WO 05/094818, US2006/0014700, US2006/0025347, WO 06/069063, WO 06/010118, WO 06/017295, and WO08/134679, all of which are incorporated herein by reference.
  • 25 BCL-2 inhibitors include but are not limited to, 4-[4-[[2-(4-Chlorophenyl)-5,5-dimethyl-1- cyclohexen-1-yl]methyl]-1-piperazinyl]-N-[[4-[[(1R)-3-(4-morpholinyl)-1- [(phenylthio)methyl]propyl]amino]-3-[(trifluoromethyl)sulfonyl]phenyl]sulfonyl]benzamide (also known as ABT-263 and described in PCT Publication No.
  • Proapoptotic receptor agonists including DR4 (TRAILR1) and DR5 (TRAILR 2 ), 35 including but are not limited to, Dulanermin (AMG-951, RhApo2L/TRAIL); Mapatumumab (HRS-ETR1, CAS 658052-09-6); Lexatumumab (HGS-ETR 2 , CAS 845816-02-6); Apomab (Apomab®); Conatumumab (AMG655, CAS 896731-82-1); and Tigatuzumab(CS1008, CAS 946415-34-5, available from Daiichi Sankyo).
  • PARAs Proapoptotic receptor agonists
  • DR4 DR4
  • DR5 DR5
  • 35 including but are not limited to, Dulanermin (AMG-951, RhApo2L/TRAIL); Mapatumumab (HRS-ETR1, CAS 658052-09-6); Lexatumumab (
  • Checkpoint Kinase (CHK) inhibitors include but are not limited to, 7-Hydroxystaurosporine (UCN- 01); 6-Bromo-3-(1-methyl-1H-pyrazol-4-yl)-5-(3R)-3-piperidinylpyrazolo[1,5-a]pyrimidin-7-amine 5 (SCH900776, CAS 891494-63-6); 5-(3-Fluorophenyl)-3-ureidothiophene-2-carboxylic acid N-[(S)- piperidin-3-yl]amide (AZD7762, CAS 860352-01-8); 4-[((3S)-1-Azabicyclo[2.2.2]oct-3-yl)amino]-3-(1H- benzimidazol-2-yl)-6-chloroquinolin-2(1H)-one (CHIR 124, CAS 405168-58-3); 7-Aminodactinomycin (7-AAD),
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, 15 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more immunomodulators (e.g., one or more of an activator of a costimulatory molecule or an inhibitor of an immune checkpoint molecule), for treating a disease, e.g., cancer..
  • immunomodulators e.g., one or more of an activator of a costimulatory molecule or an inhibitor of an immune checkpoint molecule
  • the immunomodulator is an activator of a costimulatory molecule.
  • the agonist of the costimulatory molecule is selected from an agonist (e.g., an agonistic 20 antibody or antigen-binding fragment thereof, or a soluble fusion) of OX40, CD2, CD27, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD30, CD40, BAFFR, HVEM, CD7, LIGHT, NKG2C, SLAMF7, NKp80, CD160, B7-H3 or CD83 ligand.
  • an agonist e.g., an agonistic 20 antibody or antigen-binding fragment thereof, or a soluble fusion
  • OX40 e.g., an agonistic 20 antibody or antigen-binding fragment thereof, or a soluble fusion
  • CD2 e.g., an agonistic 20 antibody or antigen-binding fragment thereof,
  • a GITR agonist is used in combination with a compound of Formula (I), or 25 a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, for treating a disease, e.g., cancer.
  • the GITR agonist is GWN323 (Novartis), BMS-986156, MK- 4166 or MK-1248 (Merck), TRX518 (Leap Therapeutics), INCAGN1876 (Incyte/Agenus), AMG 228 (Amgen) or INBRX-110 (Inhibrx).
  • GWN323 Novartis
  • BMS-986156 MK- 4166 or MK-1248
  • TRX518 Leap Therapeutics
  • INCAGN1876 Incyte/Agenus
  • AMG 228 Amgen
  • INBRX-110 Inhibrx
  • the GITR agonist is an anti-GITR antibody molecule. In one embodiment, the GITR agonist is an anti-GITR antibody molecule as described in WO 2016/057846, published on April 14, 2016, entitled“Compositions and Methods of Use for Augmented Immune Response and Cancer Therapy,” incorporated by reference in its entirety.
  • the anti-GITR antibody molecule comprises at least one, two, three, four, five 35 or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 1 (e.g., from the heavy and light chain variable region sequences of MAB7 disclosed in Table 1), or encoded by a nucleotide sequence shown in Table 1.
  • CDRs are according to the Kabat definition (e.g., as set out in Table 1).
  • the CDRs are according to the Chothia definition (e.g., as set out in Table 1).
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) 5 or deletions, relative to an amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1.
  • amino acid substitutions e.g., conservative amino acid substitutions
  • the anti-GITR antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 9, a VHCDR 2 amino acid sequence of SEQ ID NO: 11, and a VHCDR3 amino acid sequence of SEQ ID NO: 13; and a light chain variable region (VL) 10 comprising a VLCDR1 amino acid sequence of SEQ ID NO: 14, a VLCDR 2 amino acid sequence of SEQ ID NO: 16, and a VLCDR3 amino acid sequence of SEQ ID NO: 18, each disclosed in Table 1.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-GITR antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 1, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 1.
  • the anti-GITR antibody molecule comprises a VL comprising the 15 amino acid sequence of SEQ ID NO: 2, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 2.
  • the anti-GITR antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 1 and a VL comprising the amino acid sequence of SEQ ID NO: 2.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of 20 SEQ ID NO: 5, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 5. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 6, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 6. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 5 and a VL encoded by the nucleotide sequence of SEQ ID NO: 6.
  • the anti-GITR antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 3, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 3.
  • the anti-GITR antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 4, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 4.
  • the anti-GITR antibody molecule 30 comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 3 and a light chain comprising the amino acid sequence of SEQ ID NO: 4.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 7, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 7. In one embodiment, the antibody molecule comprises a light chain encoded by the 35 nucleotide sequence of SEQ ID NO: 8, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 8. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 7 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 8.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in WO 2016/057846, incorporated by reference in its entirety.
  • the anti-GITR antibody molecule is BMS-986156 (Bristol-Myers Squibb), also known as BMS 986156 or BMS986156.
  • BMS-986156 and other anti-GITR antibodies are disclosed, e.g., in US 9,228,016 and WO 2016/196792, incorporated by reference in their entirety.
  • the 5 anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BMS-986156, e.g., as disclosed in Table 2.
  • the anti-GITR antibody molecule is MK-4166 or MK-1248 (Merck).
  • MK- 4166, MK-1248, and other anti-GITR antibodies are disclosed, e.g., in US 8,709,424, WO 2011/028683, 10 WO 2015/026684, and Mahne et al. Cancer Res.2017; 77(5):1108-1118, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of MK-4166 or MK-1248.
  • the anti-GITR antibody molecule is TRX518 (Leap Therapeutics).
  • TRX518 15 and other anti-GITR antibodies are disclosed, e.g., in US 7,812,135, US 8,388,967, US 9,028,823, WO 2006/105021, and Ponte J et al. (2010) Clinical Immunology; 135:S96, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of TRX518.
  • the anti-GITR antibody molecule is INCAGN1876 (Incyte/Agenus).
  • INCAGN1876 and other anti-GITR antibodies are disclosed, e.g., in US 2015/0368349 and WO 2015/184099, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of 25 INCAGN1876.
  • the anti-GITR antibody molecule is AMG 228 (Amgen).
  • AMG 228 and other anti-GITR antibodies are disclosed, e.g., in US 9,464,139 and WO 2015/031667, incorporated by reference in their entirety.
  • the anti-GITR antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of AMG 228.
  • the anti-GITR antibody molecule is INBRX-110 (Inhibrx).
  • INBRX-110 and other anti-GITR antibodies are disclosed, e.g., in US 2017/0022284 and WO 2017/015623, incorporated 5 by reference in their entirety.
  • the GITR agonist comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of INBRX-110.
  • the GITR agonist (e.g., a fusion protein) is MEDI 1873 (MedImmune), also known as MEDI1873.
  • MEDI 1873 and other GITR agonists are disclosed, e.g., in US 2017/0073386, WO 10 2017/025610, and Ross et al. Cancer Res 2016; 76(14 Suppl): Abstract nr 561, incorporated by reference in their entirety.
  • the GITR agonist comprises one or more of an IgG Fc domain, a functional multimerization domain, and a receptor binding domain of a glucocorticoid-induced TNF receptor ligand (GITRL) of MEDI 1873.
  • GITRL glucocorticoid-induced TNF receptor ligand
  • GITR agonists include those described, e.g., in WO 15 2016/054638, incorporated by reference in its entirety.
  • the anti-GITR antibody is an antibody that competes for binding with, and/or binds to the same epitope on GITR as, one of the anti-GITR antibodies described herein.
  • the GITR agonist is a peptide that activates the GITR signaling pathway.
  • the GITR agonist is an immunoadhesin binding fragment (e.g., an immunoadhesin 20 binding fragment comprising an extracellular or GITR binding portion of GITRL) fused to a constant region (e.g., an Fc region of an immunoglobulin sequence).
  • the immunomodulator is an inhibitor of an immune checkpoint molecule. 25 In one embodiment, the immunomodulator is an inhibitor of PD-1, PD-L1, PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFRbeta. In one embodiment, the inhibitor of an immune checkpoint molecule inhibits PD-1, PD-L1, LAG-3, TIM-3 or CTLA4, or any combination thereof.
  • the term“inhibition” or“inhibitor” includes a reduction in a certain parameter, e.g., an activity, of a given molecule, e.g., an immune checkpoint inhibitor. For example, inhibition of an activity, e.g., a PD-1 or PD- L1 activity, of at least 5%, 10%, 20%, 30%, 40%, 50% or more is included by this term. Thus, inhibition need not be 100%.
  • Inhibition of an inhibitory molecule can be performed at the DNA, RNA or protein level.
  • an inhibitory nucleic acid e.g., a dsRNA, siRNA or shRNA
  • a dsRNA, siRNA or shRNA can be used to inhibit 5 expression of an inhibitory molecule.
  • the inhibitor of an inhibitory signal is a polypeptide e.g., a soluble ligand (e.g., PD-1-Ig or CTLA-4 Ig), or an antibody or antigen-binding fragment thereof, that binds to the inhibitory molecule; e.g., an antibody or fragment thereof (also referred to herein as“an antibody molecule”) that binds to PD-1, PD-L1, PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR beta, or a combination thereof.
  • a polypeptide e.g., a soluble ligand (e.g., PD-1-Ig or CTLA-4 Ig), or an antibody or antigen-binding fragment thereof, that binds to the inhibitory molecule; e.g., an antibody or fragment thereof (also referred to herein as“an antibody molecule”) that binds to PD-1, PD
  • the antibody molecule is a full antibody or fragment thereof (e.g., a Fab, F(ab')2, Fv, or a single chain Fv fragment (scFv)).
  • the antibody molecule has a heavy chain constant region (Fc) selected from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE; particularly, selected from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4, more particularly, the heavy chain constant region of IgG1 or IgG4 (e.g., human 15 IgG1 or IgG4).
  • Fc heavy chain constant region
  • the heavy chain constant region is human IgG1 or human IgG4.
  • the constant region is altered, e.g., mutated, to modify the properties of the antibody molecule (e.g., to increase or decrease one or more of Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function).
  • the antibody molecule is in the form of a bispecific or multispecific 20 antibody molecule.
  • the bispecific antibody molecule has a first binding specificity to PD-1 or PD-L1 and a second binding specificity, e.g., a second binding specificity to TIM-3, LAG-3, or PD-L2.
  • the bispecific antibody molecule binds to PD-1 or PD-L1 and TIM-3.
  • the bispecific antibody molecule binds to PD-1 or PD-L1 and LAG-3.
  • the bispecific antibody molecule binds to PD-1 and PD-L1.
  • the 25 bispecific antibody molecule binds to PD-1 and PD-L2.
  • the bispecific antibody molecule binds to TIM-3 and LAG-3.
  • Any combination of the aforesaid molecules can be made in a multispecific antibody molecule, e.g., a trispecific antibody that includes a first binding specificity to PD- 1 or PD-1, and a second and third binding specificities to two or more of TIM-3, LAG-3, or PD-L2.
  • the immunomodulator is an inhibitor of PD-1, e.g., human PD-1.
  • the immunomodulator is an inhibitor of PD-L1, e.g., human PD-L1.
  • the inhibitor of PD-1 or PD-L1 is an antibody molecule to PD-1 or PD-L1.
  • the PD-1 or PD- L1 inhibitor can be administered alone, or in combination with other immunomodulators, e.g., in combination with an inhibitor of LAG-3, TIM-3 or CTLA4.
  • the inhibitor of PD-1 or PD-L1, e.g., the anti-PD-1 or PD-L1 antibody molecule is administered in combination with a 35 LAG-3 inhibitor, e.g., an anti-LAG-3 antibody molecule.
  • the inhibitor of PD-1 or PD-L1, e.g., the anti-PD-1 or PD-L1 antibody molecule is administered in combination with a TIM-3 inhibitor, e.g., an anti-TIM-3 antibody molecule.
  • the inhibitor of PD-1 or PD- L1 is administered in combination with a LAG-3 inhibitor, e.g., an anti-LAG-3 antibody molecule, and a TIM-3 inhibitor, e.g., an anti-TIM-3 antibody molecule.
  • immunomodulators with a PD-1 inhibitor e.g., one or more of PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR
  • PD-1 inhibitor e.g., one or more of PD-L2, CTLA4, TIM3, LAG3, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4 and/or TGFR
  • Any of the antibody molecules known in the art or disclosed herein can be used in the aforesaid combinations of inhibitors of checkpoint molecule.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in 10 combination with a PD-1 inhibitor to treat a disease, e.g., cancer.
  • the PD-1 inhibitor is selected from PDR001 (Novartis), Nivolumab (Bristol-Myers Squibb), Pembrolizumab (Merck & Co), Pidilizumab (CureTech), MEDI0680 (Medimmune), REGN2810 (Regeneron), TSR-042 (Tesaro), PF- 06801591 (Pfizer), BGB-A317 (Beigene), BGB-108 (Beigene), INCSHR1210 (Incyte), or AMP-224 (Amplimmune).
  • the PD-1 inhibitor is an anti-PD-1 antibody molecule. In one embodiment, the PD-1 inhibitor is an anti-PD-1 antibody molecule as described in US 2015/0210769, published on July 30, 2015, entitled“Antibody Molecules to PD-1 and Uses Thereof,” incorporated by reference in its entirety.
  • the anti-PD-1 antibody molecule comprises at least one, two, three, four, five 20 or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 3 (e.g., from the heavy and light chain variable region sequences of BAP049-Clone-E or BAP049-Clone-B disclosed in Table 3), or encoded by a nucleotide sequence shown in Table 3.
  • the CDRs are according to the Kabat definition (e.g., as set out in Table 3).
  • the CDRs are according to the Chothia 25 definition (e.g., as set out in Table 3).
  • the CDRs are according to the combined CDR definitions of both Kabat and Chothia (e.g., as set out in Table 3).
  • the combination of Kabat and Chothia CDR of VH CDR1 comprises the amino acid sequence GYTFTTYWMH (SEQ ID NO: 213).
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) 30 or deletions, relative to an amino acid sequence shown in Table 3, or encoded by a nucleotide sequence shown in Table 3.
  • the anti-PD-1 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 22, a VHCDR 2 amino acid sequence of SEQ ID NO: 23, and a VHCDR3 amino acid sequence of SEQ ID NO: 24; and a light chain variable region (VL) 35 comprising a VLCDR1 amino acid sequence of SEQ ID NO: 31, a VLCDR 2 amino acid sequence of SEQ ID NO: 32, and a VLCDR3 amino acid sequence of SEQ ID NO: 286, each disclosed in Table 3.
  • VH heavy chain variable region
  • VL light chain variable region
  • the antibody molecule comprises a VH comprising a VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 45, a VHCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 46, and a VHCDR3 encoded by the nucleotide sequence of SEQ ID NO: 47; and a VL comprising a VLCDR1 encoded by the nucleotide sequence of SEQ ID NO: 50, a VLCDR 2 encoded by the nucleotide 5 sequence of SEQ ID NO: 51, and a VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 52, each disclosed in Table 3.
  • the anti-PD-1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 27, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 27. In one embodiment, the anti-PD-1 antibody molecule comprises a VL comprising the 10 amino acid sequence of SEQ ID NO: 41, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 41. In one embodiment, the anti-PD-1 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 37, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 37.
  • the anti-PD-1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 27 and a VL comprising the amino 15 acid sequence of SEQ ID NO: 41. In one embodiment, the anti-PD-1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 27 and a VL comprising the amino acid sequence of SEQ ID NO: 37.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 28, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID 20 NO: 28. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 42 or 38, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 42 or 38. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 28 and a VL encoded by the nucleotide sequence of SEQ ID NO: 42 or 38.
  • the anti-PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 29, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 29.
  • the anti-PD-1 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 43, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 43.
  • the anti-PD-1 antibody 30 molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 39, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 39.
  • the anti-PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 29 and a light chain comprising the amino acid sequence of SEQ ID NO: 43. In one embodiment, the anti- PD-1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 29 35 and a light chain comprising the amino acid sequence of SEQ ID NO: 39.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 30, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 30. In one embodiment, the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 44 or 40, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 44 or 40. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 30 and a light chain encoded by the nucleotide 5 sequence of SEQ ID NO: 44 or 40.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in US 2015/0210769, incorporated by reference in its entirety.
  • the anti-PD-1 antibody is Nivolumab (CAS Registry Number: 946414-94- 4).
  • Alternative names for Nivolumab include MDX-1106, MDX-1106-04, ONO-4538, BMS-936558 or OPDIVO®.
  • Nivolumab is a fully human IgG4 monoclonal antibody, which specifically blocks PD1.
  • 5 Nivolumab (clone 5C4) and other human monoclonal antibodies that specifically bind to PD1 are disclosed in US Pat No. 8,008,449 and PCT Publication No. WO2006/121168, incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Nivolumab, e.g., as disclosed in Table 4.
  • the anti-PD-1 antibody is Pembrolizumab.
  • Pembrolizumab (Trade name KEYTRUDA formerly Lambrolizumab, also known as Merck 3745, MK-3475 or SCH-900475) is a humanized IgG4 monoclonal antibody that binds to PD1.
  • Pembrolizumab is disclosed, e.g., in Hamid, O. et al.
  • the anti-PD- 1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Pembrolizumab, e.g., as disclosed in Table 4.
  • the anti-PD-1 antibody is Pidilizumab.
  • Pidilizumab (CT-011; Cure Tech) is 10 a humanized IgG1k monoclonal antibody that binds to PD1.
  • Pidilizumab and other humanized anti-PD-1 monoclonal antibodies are disclosed in PCT Publication No. WO2009/101611, incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Pidilizumab, e.g., as disclosed in Table 4.
  • anti-PD1 antibodies include AMP 514 (Amplimmune).
  • the anti-PD-1 antibody molecule is MEDI0680 (Medimmune), also known as AMP-514. MEDI0680 and other anti-PD-1 antibodies are disclosed in US 9,205,148 and WO 2012/145493, 20 incorporated by reference in their entirety.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of MEDI0680.
  • the anti-PD-1 antibody molecule is REGN2810 (Regeneron). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or 25 collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of REGN2810.
  • the anti-PD-1 antibody molecule is PF-06801591 (Pfizer). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain 30 sequence of PF-06801591.
  • the anti-PD-1 antibody molecule is BGB-A317 or BGB-108 (Beigene). In one embodiment, the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BGB-A317 or BGB-108.
  • the anti-PD-1 antibody molecule is INCSHR1210 (Incyte), also known as INCSHR01210 or SHR-1210.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of INCSHR1210.
  • the anti-PD-1 antibody molecule is TSR-042 (Tesaro), also known as ANB011.
  • the anti-PD-1 antibody molecule comprises one or more of the CDR sequences (or 5 collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of TSR-042.
  • anti-PD-1 antibodies include those described, e.g., in WO 2015/112800, WO 2016/092419, WO 2015/085847, WO 2014/179664, WO 2014/194302, WO 2014/209804, WO 2015/200119, US 8,735,553, US 7,488,802, US 8,927,697, US 8,993,731, and US 9,102,727, incorporated 10 by reference in their entirety.
  • the anti-PD-1 antibody is an antibody that competes for binding with, and/or binds to the same epitope on PD-1 as, one of the anti-PD-1 antibodies described herein.
  • the PD-1 inhibitor is a peptide that inhibits the PD-1 signaling pathway, e.g., as described in US 8,907,053, incorporated by reference in its entirety.
  • the PD-1 15 inhibitor is an immunoadhesin (e.g., an immunoadhesin comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (e.g., an Fc region of an immunoglobulin sequence).
  • the PD-1 inhibitor is AMP-224 (B7-DCIg (Amplimmune), e.g., disclosed in WO 2010/027827 and WO 2011/066342, incorporated by reference in their entirety).
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with a PD-L1 inhibitor for treating a disease, e.g., cancer.
  • a PD-L1 inhibitor for treating a disease, e.g., cancer.
  • the PD-L1 inhibitor is selected from FAZ053 (Novartis), Atezolizumab (Genentech/Roche), Avelumab (Merck Serono and Pfizer), Durvalumab (MedImmune/AstraZeneca), or BMS-936559 (Bristol-Myers Squibb).
  • the PD-L1 inhibitor is an anti-PD-L1 antibody molecule. In one embodiment, the PD-L1 inhibitor is an anti-PD-L1 antibody molecule as disclosed in US 2016/0108123, published on April 21, 2016, entitled“Antibody Molecules to PD-L1 and Uses Thereof,” incorporated by reference in its entirety.
  • the anti-PD-L1 antibody molecule comprises at least one, two, three, four, five 10 or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 5 (e.g., from the heavy and light chain variable region sequences of BAP058-Clone O or BAP058-Clone N disclosed in Table 5), or encoded by a nucleotide sequence shown in Table 5.
  • the CDRs are according to the Kabat definition (e.g., as set out in Table 5).
  • the CDRs are according to the Chothia 15 definition (e.g., as set out in Table 5).
  • the CDRs are according to the combined CDR definitions of both Kabat and Chothia (e.g., as set out in Table 5).
  • the combination of Kabat and Chothia CDR of VH CDR1 comprises the amino acid sequence GYTFTSYWMY (SEQ ID NO: 214).
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) 20 or deletions, relative to an amino acid sequence shown in Table 5, or encoded by a nucleotide sequence shown in Table 5.
  • the anti-PD-L1 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 62, a VHCDR 2 amino acid sequence of SEQ ID NO: 63, and a VHCDR3 amino acid sequence of SEQ ID NO: 64; and a light chain variable 25 region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 70, a VLCDR 2 amino acid sequence of SEQ ID NO: 71, and a VLCDR3 amino acid sequence of SEQ ID NO: 72, each disclosed in Table 5.
  • VH heavy chain variable region
  • VL light chain variable 25 region
  • the anti-PD-L1 antibody molecule comprises a VH comprising a VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 89, a VHCDR 2 encoded by the nucleotide sequence 30 of SEQ ID NO: 90, and a VHCDR3 encoded by the nucleotide sequence of SEQ ID NO: 91; and a VL comprising a VLCDR1 encoded by the nucleotide sequence of SEQ ID NO: 94, a VLCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 95, and a VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 96, each disclosed in Table 5.
  • the anti-PD-L1 antibody molecule comprises a VH comprising the amino acid 35 sequence of SEQ ID NO: 67, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 67. In one embodiment, the anti-PD-L1 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 77, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 77.
  • the anti-PD-L1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 81, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 81. In one embodiment, the anti-PD-L1 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 85, or an amino acid sequence at least 5 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 85. In one embodiment, the anti-PD-L1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 67 and a VL comprising the amino acid sequence of SEQ ID NO: 77. In one embodiment, the anti-PD-L1 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 81 and a VL comprising the amino acid sequence of SEQ ID NO: 85.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 68, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 68. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 78, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 78.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence 15 of SEQ ID NO: 82, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 82. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 86, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 86. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 68 and a VL encoded by the nucleotide sequence of SEQ ID NO: 78. In one embodiment, 20 the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 82 and a VL encoded by the nucleotide sequence of SEQ ID NO: 86.
  • the anti-PD-L1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 69, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 69. In one embodiment, the anti-PD-L1 antibody molecule comprises a 25 light chain comprising the amino acid sequence of SEQ ID NO: 79, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 79.
  • the anti-PD-L1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 83, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 83. In one embodiment, the anti-PD-L1 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID 30 NO: 87, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 87.
  • the anti-PD-L1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 69 and a light chain comprising the amino acid sequence of SEQ ID NO: 79. In one embodiment, the anti-PD-L1 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 83 and a light chain comprising the amino acid sequence of SEQ ID NO: 87. 35 In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 76, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 76.
  • the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 80, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 80. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 84, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 84.
  • the antibody molecule comprises a light chain 5 encoded by the nucleotide sequence of SEQ ID NO: 88, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 88.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 76 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 80.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 84 and a light chain encoded by the nucleotide 10 sequence of SEQ ID NO: 88.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in US 2016/0108123, incorporated by reference in its entirety.
  • the PD-L1 inhibitor is anti-PD-L1 antibody.
  • the anti- PD-L1 inhibitor is selected from YW243.55.S70, MPDL3280A, MEDI-4736, or MDX-1105MSB- 0010718C (also referred to as A09-246-2) disclosed in, e.g., WO 2013/0179174, and having a sequence 5 disclosed herein (or a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence specified).
  • the PD-L1 inhibitor is MDX-1105.
  • MDX-1105 also known as BMS-936559, is an anti-PD-L1 antibody described in PCT Publication No. WO 2007/005874.
  • the PD-L1 inhibitor is YW243.55.S70.
  • the YW243.55.S70 antibody is an 10 anti-PD-L1 described in PCT Publication No. WO 2010/077634.
  • the PD-L1 inhibitor is MDPL3280A (Genentech / Roche) also known as Atezolizumabm, RG7446, RO5541267, YW243.55.S70, or TECENTRIQTM.
  • MDPL3280A is a human Fc optimized IgG1 monoclonal antibody that binds to PD-L1.
  • MDPL3280A and other human monoclonal antibodies to PD-L1 are disclosed in U.S. Patent No.: 7,943,743 and U.S Publication No.: 20120039906 15 incorporated by reference in its entirety.
  • the anti-PD-L1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Atezolizumab, e.g., as disclosed in Table 6.
  • the PD-L2 inhibitor is AMP-224.
  • AMP-224 is a PD-L2 Fc fusion soluble 20 receptor that blocks the interaction between PD1 and B7-H1 (B7-DCIg; Amplimmune; e.g., disclosed in PCT Publication Nos. WO2010/027827 and WO2011/066342).
  • the PD-L1 inhibitor is an anti-PD-L1 antibody molecule.
  • the anti-PD-L1 antibody molecule is Avelumab (Merck Serono and Pfizer), also known as MSB0010718C. Avelumab and other anti-PD-L1 antibodies are disclosed in WO 2013/079174, incorporated by reference 25 in its entirety.
  • the anti-PD-L1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Avelumab, e.g., as disclosed in Table 6.
  • the anti-PD-L1 antibody molecule is Durvalumab (MedImmune/AstraZeneca), also known as MEDI4736. Durvalumab and other anti-PD-L1 antibodies are disclosed in US 8,779,108, 30 incorporated by reference in its entirety.
  • the anti-PD-L1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of Durvalumab, e.g., as disclosed in Table 6.
  • the anti-PD-L1 antibody molecule is BMS-936559 (Bristol-Myers Squibb), also known as MDX-1105 or 12A4.
  • the anti- PD-L1 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR 5 sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BMS-936559, e.g., as disclosed in Table 6.
  • anti-PD-L1 antibodies include those described, e.g., in WO 2015/181342, WO 2014/100079, WO 2016/000619, WO 2014/022758, WO 2014/055897, WO 2015/061668, WO 2013/079174, WO 2012/145493, WO 2015/112805, WO 2015/109124, WO 2015/195163, US 8,168,179, 10 US 8,552,154, US 8,460,927, and US 9,175,082, incorporated by reference in their entirety.
  • the anti-PD-L1 antibody is an antibody that competes for binding with, and/or binds to the same epitope on PD-L1 as, one of the anti-PD-L1 antibodies described herein.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with a LAG-3 inhibitor to treat a disease, e.g., cancer.
  • the LAG-3 inhibitor is selected from LAG525 (Novartis), BMS-986016 (Bristol-Myers Squibb), or TSR-033 (Tesaro).
  • the LAG-3 inhibitor is an anti-LAG-3 antibody molecule. In one embodiment, 5 the LAG-3 inhibitor is an anti-LAG-3 antibody molecule as disclosed in US 2015/0259420, published on September 17, 2015, entitled“Antibody Molecules to LAG-3 and Uses Thereof,” incorporated by reference in its entirety.
  • the anti-LAG-3 antibody molecule comprises at least one, two, three, four, five or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and 10 light chain variable region comprising an amino acid sequence shown in Table 7 (e.g., from the heavy and light chain variable region sequences of BAP050-Clone I or BAP050-Clone J disclosed in Table 7), or encoded by a nucleotide sequence shown in Table 7.
  • the CDRs are according to the Kabat definition (e.g., as set out in Table 7).
  • the CDRs are according to the Chothia definition (e.g., as set out in Table 7).
  • the CDRs are according to the combined CDR 15 definitions of both Kabat and Chothia (e.g., as set out in Table 7).
  • the combination of Kabat and Chothia CDR of VH CDR1 comprises the amino acid sequence GFTLTNYGMN (SEQ ID NO: 173).
  • one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) or deletions, relative to an amino acid sequence shown in Table 7, or encoded by a nucleotide sequence 20 shown in Table 7.
  • the anti-LAG-3 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 108, a VHCDR 2 amino acid sequence of SEQ ID NO: 109, and a VHCDR3 amino acid sequence of SEQ ID NO: 110; and a light chain variable region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 117, a VLCDR 2 amino acid 25 sequence of SEQ ID NO: 118, and a VLCDR3 amino acid sequence of SEQ ID NO: 119, each disclosed in Table 7.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-LAG-3 antibody molecule comprises a VH comprising a VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 143 or 144, a VHCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 145 or 146, and a VHCDR3 encoded by the nucleotide sequence of SEQ ID NO: 30 147 or 148; and a VL comprising a VLCDR1 encoded by the nucleotide sequence of SEQ ID NO: 153 or 154, a VLCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 155 or 156, and a VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 157 or 158, each disclosed in Table 7.
  • the anti-LAG-3 antibody molecule comprises a VH comprising a VHCDR1 encoded by the nucleotide sequence of SEQ ID NO: 165 or 144, a VHCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 166 35 or 146, and a VHCDR3 encoded by the nucleotide sequence of SEQ ID NO: 167 or 148; and a VL comprising a VLCDR1 encoded by the nucleotide sequence of SEQ ID NO: 153 or 154, a VLCDR 2 encoded by the nucleotide sequence of SEQ ID NO: 155 or 156, and a VLCDR3 encoded by the nucleotide sequence of SEQ ID NO: 157 or 158, each disclosed in Table 7.
  • the anti-LAG-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 113, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or 5 higher to SEQ ID NO: 113. In one embodiment, the anti-LAG-3 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 125, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 125. In one embodiment, the anti-LAG-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 131, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 131.
  • the 10 anti-LAG-3 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 137, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 137.
  • the anti-LAG-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 113 and a VL comprising the amino acid sequence of SEQ ID NO: 125.
  • the anti-LAG-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 15 131 and a VL comprising the amino acid sequence of SEQ ID NO: 137.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 114 or 115, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 114 or 115. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 126 or 127, or a nucleotide sequence at least 85%, 90%, 95%, or 99% 20 identical or higher to SEQ ID NO: 126 or 127.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 132 or 133, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 132 or 133.
  • the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 138 or 139, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 138 or 139.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO:
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 132 or 133 and a VL encoded by the nucleotide sequence of SEQ ID NO: 138 or 139.
  • the anti-LAG-3 antibody molecule comprises a heavy chain comprising the 30 amino acid sequence of SEQ ID NO: 116, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 116.
  • the anti-LAG-3 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 128, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 128.
  • the anti-LAG-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 134, or 35 an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 134.
  • the anti-LAG-3 antibody molecule comprises a light chain comprising the amino acid sequence of SEQ ID NO: 140, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 140.
  • the anti-LAG-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 116 and a light chain comprising the amino acid sequence of SEQ ID NO: 128.
  • the anti-LAG-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 134 and a light chain comprising the amino acid 5 sequence of SEQ ID NO: 140.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 123 or 124, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 123 or 124. In one embodiment, the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 129 or 130, or a nucleotide sequence at least 85%, 10 90%, 95%, or 99% identical or higher to SEQ ID NO: 129 or 130.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 135 or 136, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 135 or 136. In one embodiment, the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 141 or 142, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ 15 ID NO: 141 or 142.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 123 or 124 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 129 or 130. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 135 or 136 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 141 or 142.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described in US 2015/0259420, incorporated by reference in its entirety.
  • the anti-LAG-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of BMS-986016, e.g., as disclosed in Table 8. 5
  • the anti-LAG-3 antibody molecule is TSR-033 (Tesaro).
  • the anti-LAG-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of TSR-033.
  • the anti-LAG-3 antibody molecule is IMP731 or GSK2831781 (GSK and 10 Prima BioMed). IMP731 and other anti-LAG-3 antibodies are disclosed in WO 2008/132601 and US 9,244,059, incorporated by reference in their entirety.
  • the anti-LAG-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of IMP731, e.g., as disclosed in Table 8.
  • the anti-LAG-3 antibody molecule comprises one or more 15 of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of GSK2831781.
  • the anti-LAG-3 antibody molecule is IMP761 (Prima BioMed). In one embodiment, the anti-LAG-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy 20 chain or light chain sequence of IMP761.
  • anti-LAG-3 antibodies include those described, e.g., in WO 2008/132601, WO 2010/019570, WO 2014/140180, WO 2015/116539, WO 2015/200119, WO 2016/028672, US 9,244,059, US 9,505,839, incorporated by reference in their entirety.
  • the anti-LAG-3 antibody is an antibody that competes for binding with, and/or 25 binds to the same epitope on LAG-3 as, one of the anti-LAG-3 antibodies described herein.
  • the anti-LAG-3 inhibitor is a soluble LAG-3 protein, e.g., IMP321 (Prima BioMed), e.g., as disclosed in WO 2009/044273, incorporated by reference in its entirety.
  • IMP321 Primary BioMed
  • the inhibitor of an immune checkpoint molecule is an inhibitor of TIM-3.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with 5 a TIM-3 inhibitor to treat a disease, e.g., cancer.
  • the TIM-3 inhibitor is MGB453 (Novartis) or TSR-022 (Tesaro).
  • the TIM-3 inhibitor is an anti-TIM-3 antibody molecule. In one embodiment, the TIM-3 inhibitor is an anti-TIM-3 antibody molecule as disclosed in US 2015/0218274, published on 10 August 6, 2015, entitled“Antibody Molecules to TIM-3 and Uses Thereof,” incorporated by reference in its entirety.
  • the anti-TIM-3 antibody molecule comprises at least one, two, three, four, five or six complementarity determining regions (CDRs) (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 9 (e.g., from the heavy and light chain variable region sequences of ABTIM3-hum11 or ABTIM3-hum03 disclosed in Table 9), or encoded by a nucleotide sequence shown in Table 9.
  • the CDRs are according to the Kabat definition (e.g., as set out in Table 9).
  • the CDRs are according to the Chothia definition (e.g., as set out in Table 9).
  • one or more of the CDRs (or collectively all of 5 the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions (e.g., conservative amino acid substitutions) or deletions, relative to an amino acid sequence shown in Table 9, or encoded by a nucleotide sequence shown in Table 9.
  • amino acid substitutions e.g., conservative amino acid substitutions
  • deletions e.g., conservative amino acid substitutions
  • the anti-TIM-3 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 174, a VHCDR 2 amino acid sequence 10 of SEQ ID NO: 175, and a VHCDR3 amino acid sequence of SEQ ID NO: 176; and a light chain variable region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 183, a VLCDR 2 amino acid sequence of SEQ ID NO: 184, and a VLCDR3 amino acid sequence of SEQ ID NO: 185, each disclosed in Table 9.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-TIM-3 antibody molecule comprises a heavy chain variable region (VH) comprising a VHCDR1 amino acid sequence of SEQ ID NO: 174, a VHCDR 2 amino acid sequence 15 of SEQ ID NO: 193, and a VHCDR3 amino acid sequence of SEQ ID NO: 176; and a light chain variable region (VL) comprising a VLCDR1 amino acid sequence of SEQ ID NO: 183, a VLCDR 2 amino acid sequence of SEQ ID NO: 184, and a VLCDR3 amino acid sequence of SEQ ID NO: 185, each disclosed in Table 9.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-TIM-3 antibody molecule comprises a VH comprising the amino acid 20 sequence of SEQ ID NO: 179, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 179. In one embodiment, the anti-TIM-3 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 189, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 189. In one embodiment, the anti-TIM-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 195, or an amino acid sequence at 25 least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 195.
  • the anti-TIM-3 antibody molecule comprises a VL comprising the amino acid sequence of SEQ ID NO: 199, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 199.
  • the anti-TIM-3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 179 and a VL comprising the amino acid sequence of SEQ ID NO: 189.
  • the anti-TIM- 30 3 antibody molecule comprises a VH comprising the amino acid sequence of SEQ ID NO: 195 and a VL comprising the amino acid sequence of SEQ ID NO: 199.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 180, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 180. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence 35 of SEQ ID NO: 190, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 190.
  • the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 196, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 196. In one embodiment, the antibody molecule comprises a VL encoded by the nucleotide sequence of SEQ ID NO: 200, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 200. In one embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO: 180 and a VL encoded by the nucleotide sequence of SEQ ID NO: 190. In one 5 embodiment, the antibody molecule comprises a VH encoded by the nucleotide sequence of SEQ ID NO:
  • the anti-TIM-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 181, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 181.
  • the anti-TIM-3 antibody molecule comprises 10 a light chain comprising the amino acid sequence of SEQ ID NO: 191, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 191.
  • the anti-TIM-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 197, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 197.
  • the anti-TIM-3 antibody molecule comprises a light chain comprising the amino acid 15 sequence of SEQ ID NO: 201, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 201.
  • the anti-TIM-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 181 and a light chain comprising the amino acid sequence of SEQ ID NO: 191.
  • the anti-TIM-3 antibody molecule comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 197 and a light chain comprising the amino acid 20 sequence of SEQ ID NO: 201.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 182, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 182.
  • the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 192, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical 25 or higher to SEQ ID NO: 192.
  • the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 198, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 198. In one embodiment, the antibody molecule comprises a light chain encoded by the nucleotide sequence of SEQ ID NO: 202, or a nucleotide sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 202. In one embodiment, the antibody molecule comprises a 30 heavy chain encoded by the nucleotide sequence of SEQ ID NO: 182 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 192. In one embodiment, the antibody molecule comprises a heavy chain encoded by the nucleotide sequence of SEQ ID NO: 198 and a light chain encoded by the nucleotide sequence of SEQ ID NO: 202.
  • the antibody molecules described herein can be made by vectors, host cells, and methods described 35 in US 2015/0218274, incorporated by reference in its entirety.
  • the anti-TIM-3 antibody molecule is TSR-022 (AnaptysBio/Tesaro). In one embodiment, the anti-TIM-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy 5 chain or light chain sequence of TSR-022. In one embodiment, the anti-TIM-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of APE5137 or APE5121, e.g., as disclosed in Table 10. APE5137, APE5121, and other anti-TIM-3 antibodies are disclosed in WO 2016/161270, incorporated by reference in its entirety.
  • the anti-TIM-3 antibody molecule is the antibody clone F38-2E2. In one embodiment, the anti-TIM-3 antibody molecule comprises one or more of the CDR sequences (or collectively all of the CDR sequences), the heavy chain or light chain variable region sequence, or the heavy chain or light chain sequence of F38-2E2.
  • anti-TIM-3 antibodies include those described, e.g., in WO 2016/111947, WO 15 2016/071448, WO 2016/144803, US 8,552,156, US 8,841,418, and US 9,163,087, incorporated by reference in their entirety.
  • the anti-TIM-3 antibody is an antibody that competes for binding with, and/or binds to the same epitope on TIM-3 as, one of the anti-TIM-3 antibodies described herein.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more cytokines, including but not limited to, interferon, IL-2, IL-15, IL-7, or IL21. 5
  • compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof are administered in combination with an IL-15/IL-15Ra complex.
  • the IL-15/IL-15Ra complex is selected from NIZ985 (Novartis), ATL-803 (Altor) or CYP0150 (Cytune).
  • the cytokine is IL-15 complexed with a soluble form of IL-15 receptor alpha (IL-15Ra).
  • the IL-15/IL-15Ra complex may comprise IL-15 covalently or noncovalently bound to a soluble form of IL-15Ra.
  • the human IL-15 is noncovalently bonded to a soluble form of IL-15Ra.
  • the human IL-15 of the formulation comprises an amino acid sequence of SEQ ID NO: 207 in Table 11 or an amino acid sequence at least 85%, 90%, 95%, or 99% 15 identical or higher to SEQ ID NO: 207
  • the soluble form of human IL-15Ra comprises an amino acid sequence of SEQ ID NO: 208 in Table 11, or an amino acid sequence at least 85%, 90%, 95%, or 99% identical or higher to SEQ ID NO: 208, as described in WO 2014/066527, incorporated by reference in its entirety.
  • the molecules described herein can be made by vectors, host cells, and methods described in WO 2007084342, incorporated by reference in its entirety.
  • the IL-15/IL-15Ra complex is ALT-803, an IL-15/IL-15Ra Fc fusion protein (IL-15N72D:IL-15RaSu/Fc soluble complex).
  • ALT-803 is described in WO 2008/143794, incorporated by reference in its entirety.
  • the IL-15/IL-15Ra Fc fusion protein comprises the sequences 5 as disclosed in Table 12.
  • the IL-15/IL-15Ra complex comprises IL-15 fused to the sushi domain of IL- 15Ra (CYP0150, Cytune).
  • the sushi domain of IL-15Ra refers to a domain beginning at the first cysteine residue after the signal peptide of IL-15Ra, and ending at the fourth cysteine residue after said signal peptide.
  • the complex of IL-15 fused to the sushi domain of IL-15Ra is described in WO 2007/04606 and WO 10 2012/175222, incorporated by reference in their entirety.
  • the IL-15/IL-15Ra sushi domain fusion comprises the sequences as disclosed in Table 12.
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more agonists of toll like receptors (TLRs, e.g., TLR7, TLR8, TLR9) to treat a disease, e.g., cancer.
  • TLRs toll like receptors
  • a compound of the present disclosure can be used in 5 combination with a TLR7 agonist or a TLR7 agonist conjugate.
  • the TLR7 agonist comprises a compound disclosed in International Application Publication No. WO2011/049677, which is hereby incorporated by reference in its entirety.
  • the TLR7 agonist comprises 3-(5-amino-2-(4-(2-(3,3-difluoro-3- phosphonopropoxy)ethoxy)-2-methylphenethyl)benzo[f][1,7]naphthyridin-8-yl)propanoic acid.
  • the TLR7 agonist comprises a compound of formula:
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more angiogenesis inhibitors to treat cancer, e.g., Bevacizumab (Avastin®),15 axitinib (Inlyta®); Brivanib alaninate (BMS-582664, (S)-((R)-1-(4-(4-Fluoro-2-methyl-1H-indol-5-yloxy)- 5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate); Sorafenib (Nexavar®); Pazopanib (Votrient®); Sunitinib malate (Sutent®); Cediranib (AZD2171, CAS 288383-20-1); Vargatef (BIBF1120
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in 5 combination with one or more heat shock protein inhibitors to treat cancer, e.g., Tanespimycin (17- allylamino-17-demethoxygeldanamycin, also known as KOS-953 and 17-AAG, available from SIGMA, and described in US Patent No.4,261,989); Retaspimycin (IPI504), Ganetespib (STA-9090); [6-Chloro-9- (4-methoxy-3,5-dimethylpyridin-2-ylmethyl)-9H-purin-2-yl]amine (BIIB021 or CNF2024, CAS 848695- 25-0); trans-4-[[2-(Aminocarbonyl)-5-[4,5,6,7-tetrahydro-6,6-dimethyl-4-oxo-3-(trifluoro
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, 15 hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more HDAC inhibitors or other epigenetic modifiers.
  • HDAC inhibitors include, but not limited to, Voninostat (Zolinza®); Romidepsin (Istodax®); Treichostatin A (TSA); Oxamflatin; Vorinostat (Zolinza®, Suberoylanilide hydroxamic acid); Pyroxamide (syberoyl-3- aminopyridineamide hydroxamic acid); Trapoxin A (RF-1023A); Trapoxin B (RF-10238); Cyclo[(aS,2S)-20 a-amino-h-oxo-2-oxiraneoctanoyl-O-methyl-D-tyrosyl-L-isoleucyl-L-prolyl] (Cyl-1); Cyclo[(aS,2S)-a- amino-h-oxo-2-oxiraneoctanoyl-O-methyl-D-tyrosyl-L-isoleucyl-(2S)-2-piperidinecarbonyl] (C
  • epigenetic modifiers include but not limited to inhibitors of EZH2 (enhancer of zeste homolog 2), EED (embryonic ectoderm development), or LSD1 (lysine-specific histone demethylase 1A or KDM1A).
  • the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof, of the present disclosure are used in combination with one or more inhibitors of indoleamine-pyrrole 2,3-dioxygenase (IDO), for example, Indoximod (also known as NLG-8189), a-Cyclohexyl-5H-imidazo[5,1-a]isoindole-5-ethanol (also known 5 as NLG919), or (4E)-4-[(3-Chloro-4-fluoroanilino)-nitrosomethylidene]-1,2,5-oxadiazol-3-amine (also known as INCB024360), to treat cancer.
  • IDO indoleamine-pyrrole 2,3-dioxygenase
  • the present disclosure provides for the compounds of Formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, prodrug, stereoisomer, or tautomer thereof for use in combination with adoptive 10 immunotherapy methods and reagents such as chimeric antigen receptor (CAR) immune effector cells, e.g., T cells, or chimeric TCR-transduced immune effector cells, e.g., T cells.
  • CAR chimeric antigen receptor
  • aspects of the present disclosure pertain to or include an isolated nucleic acid molecule encoding a chimeric antigen receptor (CAR), wherein the CAR comprises an antigen binding domain (e.g., antibody or antibody fragment, TCR or TCR fragment) that binds to a tumor antigen as described herein, a transmembrane domain (e.g., a transmembrane domain described herein), and an intracellular signaling domain (e.g., an intracellular signaling domain described herein) (e.g., an intracellular signaling domain 20 comprising a costimulatory domain (e.g., a costimulatory domain described herein) and/or a primary signaling domain (e.g., a primary signaling domain described herein).
  • an antigen binding domain e.g., antibody or antibody fragment, TCR or TCR fragment
  • TCR or TCR fragment binds to a tumor antigen as described herein
  • a transmembrane domain e.g., a transme
  • the present disclosure includes: host cells containing the above nucleic acids and isolated proteins encoded by such nucleic acid molecules.
  • CAR nucleic acid constructs, encoded proteins, containing vectors, host cells, pharmaceutical compositions, and methods of administration and treatment related to the present disclosure 25 are disclosed in detail in International Patent Application Publication No. WO2015142675, which is incorporated by reference in its entirety.
  • the disclosure pertains to an isolated nucleic acid molecule encoding a chimeric antigen receptor (CAR), wherein the CAR comprises an antigen binding domain (e.g., antibody or antibody fragment, TCR or TCR fragment) that binds to a tumor-supporting antigen (e.g., a tumor-supporting antigen 30 as described herein), a transmembrane domain (e.g., a transmembrane domain described herein), and an intracellular signaling domain (e.g., an intracellular signaling domain described herein) (e.g., an intracellular signaling domain comprising a costimulatory domain (e.g., a costimulatory domain described herein) and/or a primary signaling domain (e.g., a primary signaling domain described herein).
  • an antigen binding domain e.g., antibody or antibody fragment, TCR or TCR fragment
  • a tumor-supporting antigen e.g., a tumor-supporting antigen 30 as described here
  • the tumor-supporting antigen is an antigen present on a stromal cell or a myeloid-derived 35 suppressor cell (MDSC).
  • the disclosure features polypeptides encoded by such nucleic acids and host cells containing such nucleic acids and/or polypeptides.
  • aspects of the disclosure pertain to isolated nucleic acid encoding a chimeric T cell receptor (TCR) comprising a TCR alpha and/or TCR beta variable domain with specificity for a cancer antigen described herein. See for example, Dembic et al., Nature, 320, 232-238 (1986), Schumacher, Nat. Rev. Immunol., 2, 512-519 (2002), Kershaw et al., Nat. Rev.
  • Such chimeric TCRs may recognize, for 10 example, cancer antigens such as MART-1, gp-100, p53, and NY-ESO-1, MAGE A3/A6, MAGEA3, SSX2, HPV-16 E6 or HPV-16 E7.
  • cancer antigens such as MART-1, gp-100, p53, and NY-ESO-1, MAGE A3/A6, MAGEA3, SSX2, HPV-16 E6 or HPV-16 E7.
  • the disclosure features polypeptides encoded by such nucleic acids and host cells containing such nucleic acids and/or polypeptides.
  • the present disclosure provides cells, e.g., immune effector cells (e.g., T cells, NK cells), that comprise or at any time comprised a gRNA molecule or CRISPR system as described herein, that are further engineered to contain one or more CARs that direct the immune effector cells to undesired cells (e.g., cancer 5 cells). This is achieved through an antigen binding domain on the CAR that is specific for a cancer associated antigen.
  • immune effector cells e.g., T cells, NK cells
  • CRISPR system as described herein
  • cancer associated antigens There are two classes of cancer associated antigens (tumor antigens) that can be targeted by the CARs of the instant disclosure: (1) cancer associated antigens that are expressed on the surface of cancer cells; and (2) cancer associated antigens that itself is intracellular, however, a fragment of such antigen (peptide) is presented on the surface of the cancer cells by MHC (major histocompatibility complex). 10
  • the tumor antigen is chosen from one or more of: CD19; CD123; CD22;
  • CD30 CD171; CS-1 (also referred to as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24); C-type lectin-like molecule-1 (CLL-1 or CLECL1); CD33; epidermal growth factor receptor variant III (EGFRvIII); ganglioside G2 (GD2); ganglioside GD3 (aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(1- 4)bDGlcp(1-1)Cer); TNF receptor family member B cell maturation (BCMA); Tn antigen ((Tn Ag) or (GalNAca-Ser/Thr)); prostate-specific membrane antigen (PSMA); Receptor tyrosine kinase-like orphan receptor 1 (ROR1); Fms-Like Tyrosine Kinase 3 (FLT3); Tumor-associated glycoprotein 72 (TAG72); 5 CD38; CD44v6; Carcinoembryonic antigen (CEA
  • a CAR described herein can comprise an antigen binding domain (e.g., antibody or antibody 15 fragment, TCR or TCR fragment) that binds to a tumor-supporting antigen (e.g., a tumor-supporting antigen as described herein).
  • the tumor-supporting antigen is an antigen present on a stromal cell or a myeloid-derived suppressor cell (MDSC).
  • Stromal cells can secrete growth factors to promote cell division in the microenvironment. MDSC cells can inhibit T cell proliferation and activation.
  • the CAR-expressing cells destroy the tumor- 20 supporting cells, thereby indirectly inhibiting tumor growth or survival.
  • the stromal cell antigen is chosen from one or more of: bone marrow stromal cell antigen 2 (BST2), fibroblast activation protein (FAP) and tenascin.
  • BST2 bone marrow stromal cell antigen 2
  • FAP fibroblast activation protein
  • tenascin tenascin.
  • the FAP-specific antibody is, competes for binding with, or has the same CDRs as, sibrotuzumab.
  • the MDSC antigen is chosen from one or more of: CD33, CD11b, C14, CD15, and CD66b.
  • the tumor-supporting antigen is chosen from one or more of: bone marrow stromal cell antigen 2 (BST2), fibroblast activation protein (FAP) or tenascin, CD33, CD11b, C14, CD15, and CD66b.
  • BST2 bone marrow stromal cell antigen 2
  • FAP fibroblast activation protein
  • tenascin CD33, CD11b, C14, CD15, and CD66b.
  • the antigen binding domain of the encoded CAR molecule comprises an antibody, an antibody fragment, an scFv, a Fv, a Fab, a (Fab’)2, a single domain antibody (SDAB), a VH 30 or VL domain, a camelid VHH domain or a bi-functional (e.g. bi-specific) hybrid antibody (e.g., Lanzavecchia et al., Eur. J. Immunol.17, 105 (1987)).
  • scFvs can be prepared according to method known in the art (see, for example, Bird et al., (1988) Science 242:423-426 and Huston et al., (1988) Proc. Natl. Acad. Sci. USA 85:5879- 5883).
  • ScFv molecules can be produced by linking VH and VL regions together using flexible polypeptide 35 linkers.
  • the scFv molecules comprise a linker (e.g., a Ser-Gly linker) with an optimized length and/or amino acid composition. The linker length can greatly affect how the variable regions of a scFv fold and interact.
  • a short polypeptide linker e.g., between 5-10 amino acids
  • intrachain folding is prevented.
  • Interchain folding is also required to bring the two variable regions together to form a functional epitope binding site.
  • linker orientation and size see, e.g., Hollinger et al.1993 Proc Natl Acad. Sci. U.S.A.90:6444-6448, U.S. Patent Application Publication Nos.2005/0100543, 2005/0175606, 2007/0014794, and PCT publication Nos. WO2006/020258 and WO2007/024715, is incorporated herein 5 by reference.
  • An scFv can comprise a linker of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more amino acid residues between its VL and VH regions.
  • the linker sequence may comprise any naturally occurring amino acid.
  • the linker sequence comprises amino acids glycine and serine.
  • the linker sequence comprises sets of 10 glycine and serine repeats such as (Gly4Ser)n, where n is a positive integer equal to or greater than 1 (SEQ ID NO: 217).
  • the linker can be (Gly 4 Ser) 4 (SEQ ID NO: 215) or (Gly 4 Ser) 3 (SEQ ID NO: 216). Variation in the linker length may retain or enhance activity, giving rise to superior efficacy in activity studies.
  • the antigen binding domain is a T cell receptor (“TCR”), or a fragment thereof, 15 for example, a single chain TCR (scTCR).
  • TCR T cell receptor
  • scTCR single chain TCR
  • Methods to make such TCRs are known in the art. See, e.g., Willemsen RA et al, Gene Therapy 7: 1369–1377 (2000); Zhang T et al, Cancer Gene Ther 11: 487–496 (2004); Aggen et al, Gene Ther.19(4):365-74 (2012) (references are incorporated herein by its entirety).
  • scTCR can be engineered that contains the Va and Vb genes from a T cell clone linked by a linker (e.g., a flexible peptide). This approach is very useful to cancer associated target that itself is 20 intracellular, however, a fragment of such antigen (peptide) is presented on the surface of the cancer cells by MHC.
  • the encoded antigen binding domain has a binding affinity KD of 10 -4 M to 10 -8 M.
  • the encoded CAR molecule comprises an antigen binding domain that has a 25 binding affinity KD of 10 -4 M to 10 -8 M, e.g., 10 -5 M to 10 -7 M, e.g., 10 -6 M or 10 -7 M, for the target antigen.
  • the antigen binding domain has a binding affinity that is at least five-fold, 10-fold, 20- fold, 30-fold, 50-fold, 100-fold or 1,000-fold less than a reference antibody, e.g., an antibody described herein.
  • the encoded antigen binding domain has a binding affinity at least 5-fold less than a reference antibody (e.g., an antibody from which the antigen binding domain is derived).
  • such antibody fragments are functional in that they provide a biological response that can include, but is not limited to, activation of an immune response, inhibition of signal-transduction origination from its target antigen, inhibition of kinase activity, and the like, as will be understood by a skilled artisan.
  • the antigen binding domain of the CAR is a scFv antibody fragment that is humanized compared to the murine sequence of the scFv from which it is derived.
  • the antigen binding domain of a CAR of the disclosure is encoded by a nucleic acid molecule whose sequence has been codon optimized for expression in a mammalian cell.
  • entire CAR construct of the disclosure is encoded by a nucleic acid molecule whose entire sequence has been codon optimized for expression in a mammalian cell. Codon optimization refers to the discovery that the frequency of occurrence of synonymous codons (i.e., codons that code for the same amino acid) in coding DNA is biased in different species. Such codon degeneracy allows an identical polypeptide to be encoded by a variety of nucleotide sequences.
  • a variety of codon optimization methods 5 is known in the art, and include, e.g., methods disclosed in at least US Patent Nos 5,786,464 and 6,114,148.
  • Antigen binding domains (and the targeted antigens)
  • an antigen binding domain against CD19 is an antigen binding portion, e.g., CDRs, of a CAR, antibody or antigen-binding fragment thereof described in, e.g., PCT publication WO2012/079000; PCT publication WO2014/153270; Kochenderfer, J.N. et al., J. Immunother.32 (7), 689- 10 702 (2009); Kochenderfer, J.N., et al., Blood, 116 (20), 4099-4102 (2010); PCT publication WO2014/031687; Bejcek, Cancer Research, 55, 2346-2351, 1995; or U.S. Patent No.7,446,190.
  • an antigen binding domain against mesothelin is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment or CAR described in, e.g., PCT publication WO2015/090230.
  • an antigen binding domain against mesothelin is an antigen binding 15 portion, e.g., CDRs, of an antibody, antigen-binding fragment, or CAR described in, e.g., PCT publication WO1997/025068, WO1999/028471, WO2005/014652, WO2006/099141, WO2009/045957, WO2009/068204, WO2013/142034, WO2013/040557, or WO2013/063419.
  • an antigen binding domain against mesothelin is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment, or CAR described in WO/2015/090230.
  • an antigen binding domain against CD123 is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment or CAR described in, e.g., PCT publication WO2014/130635.
  • an antigen binding domain against CD123 is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment, or CAR described in, e.g., PCT publication WO2014/138805, WO2014/138819, WO2013/173820, WO2014/144622, WO2001/66139, 25 WO2010/126066, WO2014/144622, or US2009/0252742.
  • an antigen binding domain against CD123 is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment, or CAR described in WO/2017/028896.
  • an antigen binding domain against EGFRvIII is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment or CAR described in, e.g., WO/2014/130657.
  • an antigen binding domain against CD22 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Haso et al., Blood, 121(7): 1165-1174 (2013); Wayne et al., Clin Cancer Res 16(6): 1894-1903 (2010); Kato et al., Leuk Res 37(1):83-88 (2013); Creative BioMart (creativebiomart.net): MOM-18047-S(P).
  • an antigen binding domain against CS-1 is an antigen binding portion, e.g., 35 CDRs, of Elotuzumab (BMS), see e.g., Tai et al., 2008, Blood 112(4):1329-37; Tai et al., 2007, Blood.
  • BMS Elotuzumab
  • an antigen binding domain against CLL-1 is an antigen binding portion, e.g., CDRs, of an antibody available from R&D, ebiosciences, Abcam, for example, PE-CLL1-hu Cat# 353604 (BioLegend); and PE-CLL1 (CLEC12A) Cat# 562566 (BD).
  • an antigen binding domain against CLL-1 is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment, 5 or CAR described in WO/2017/014535.
  • an antigen binding domain against CD33 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Bross et al., Clin Cancer Res 7(6):1490-1496 (2001) (Gemtuzumab Ozogamicin, hP67.6),Caron et al., Cancer Res 52(24):6761-6767 (1992) (Lintuzumab, HuM195), Lapusan et al., Invest New Drugs 30(3):1121-1131 (2012) (AVE9633), Aigner et al., Leukemia 27(5): 1107-1115 10 (2013) (AMG330, CD33 BiTE), Dutour et al., Adv hematol 2012:683065 (2012), and Pizzitola et al., Leukemia doi:10.1038/Lue.2014.62 (2014).
  • an antigen binding domain against CD33 is an antigen binding portion, e.g., CDRs, of
  • an antigen binding domain against GD2 is an antigen binding portion, e.g., 15 CDRs, of an antibody described in, e.g., Mujoo et al., Cancer Res.47(4):1098-1104 (1987); Cheung et al., Cancer Res 45(6):2642-2649 (1985), Cheung et al., J Clin Oncol 5(9):1430-1440 (1987), Cheung et al., J Clin Oncol 16(9):3053-3060 (1998), Handgretinger et al., Cancer Immunol Immunother 35(3):199-204 (1992).
  • an antigen binding domain against GD2 is an antigen binding portion of an antibody selected from mAb 14.18, 14G2a, ch14.18, hu14.18, 3F8, hu3F8, 3G6, 8B6, 60C3, 10B8, ME36.1, 20 and 8H9, see e.g., WO2012033885, WO2013040371, WO2013192294, WO2013061273, WO2013123061, WO2013074916, and WO201385552.
  • an antigen binding domain against GD2 is an antigen binding portion of an antibody described in US Publication No.: 20100150910 or PCT Publication No.: WO 2011160119.
  • an antigen binding domain against BCMA is an antigen binding portion, e.g., 25 CDRs, of an antibody described in, e.g., WO2012163805, WO200112812, and WO2003062401.
  • an antigen binding domain against BCMA is an antigen binding portion, e.g., CDRs, of an antibody, antigen-binding fragment, or CAR described in WO/2017/014565.
  • an antigen binding domain against Tn antigen is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 8,440,798, Brooks et al., PNAS 107(22):10056-10061 30 (2010), and Stone et al., OncoImmunology 1(6):863-873(2012).
  • an antigen binding domain against PSMA is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Parker et al., Protein Expr Purif 89(2):136-145 (2013), US 20110268656 (J591 ScFv); Frigerio et al, European J Cancer 49(9):2223-2232 (2013) (scFvD2B); WO 2006125481 (mAbs 3/A12, 3/E7 and 3/F11) and single chain antibody fragments (scFv A5 and D7).
  • CDRs antigen binding portion
  • an antigen binding domain against ROR1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Hudecek et al., Clin Cancer Res 19(12):3153-3164 (2013); WO 2011159847; and US20130101607.
  • an antigen binding domain against FLT3 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., WO2011076922, US 5777084, EP0754230, US20090297529, and several commercial catalog antibodies (R&D, ebiosciences, Abcam).
  • an antigen binding domain against TAG72 is an antigen binding portion, e.g., 5 CDRs, of an antibody described in, e.g., Hombach et al., Gastroenterology 113(4):1163-1170 (1997); and Abcam ab691.
  • an antigen binding domain against FAP is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Ostermann et al., Clinical Cancer Research 14:4584-4592 (2008) (FAP5), US Pat. Publication No. 2009/0304718; sibrotuzumab (see e.g., Hofheinz et al., Oncology 10 Research and Treatment 26(1), 2003); and Tran et al., J Exp Med 210(6):1125-1135 (2013).
  • CDRs antigen binding portion
  • an antigen binding domain against CD38 is an antigen binding portion, e.g., CDRs, of daratumumab (see, e.g., Groen et al., Blood 116(21):1261-1262 (2010); MOR 2 02 (see, e.g., US 8,263,746); or antibodies described in US 8,362,211.
  • CDRs antigen binding portion
  • an antigen binding domain against CD44v6 is an antigen binding portion, e.g., 15 CDRs, of an antibody described in, e.g., Casucci et al., Blood 122(20):3461-3472 (2013).
  • an antigen binding domain against CEA is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Chmielewski et al., Gastoenterology 143(4):1095-1107 (2012).
  • an antigen binding domain against EPCAM is an antigen binding portion, e.g., CDRS, of an antibody selected from MT110, EpCAM-CD3 bispecific Ab (see, e.g., 20 clinicaltrials.gov/ct2/show/NCT00635596); Edrecolomab; 3622W94; ING-1; and adecatumumab (MT201).
  • CDRS antigen binding portion
  • EpCAM-CD3 bispecific Ab see, e.g., 20 clinicaltrials.gov/ct2/show/NCT00635596
  • Edrecolomab 3622W94
  • ING-1 adecatumumab
  • an antigen binding domain against PRSS21 is an antigen binding portion, e.g., CDRs, of an antibody described in US Patent No.: 8,080,650.
  • an antigen binding domain against B7H3 is an antigen binding portion, e.g., CDRs, of an antibody MGA271 (Macrogenics).
  • an antigen binding domain against KIT is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 7915391, US20120288506, and several commercial catalog antibodies.
  • an antigen binding domain against IL-13Ra2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., WO2008/146911, WO2004087758, several commercial 30 catalog antibodies, and WO2004087758.
  • an antigen binding domain against CD30 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 7090843 B1, and EP0805871.
  • an antigen binding domain against GD3 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 7253263; US 8,207,308; US 20120276046; EP1013761; 35 WO2005035577; and US 6437098.
  • an antigen binding domain against CD171 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Hong et al., J Immunother 37(2):93-104 (2014).
  • an antigen binding domain against IL-11Ra is an antigen binding portion, e.g., CDRs, of an antibody available from Abcam (cat# ab55262) or Novus Biologicals (cat# EPR5446).
  • an antigen binding domain again IL-11Ra is a peptide, see, e.g., Huang et al., Cancer Res 72(1):271-281 (2012).
  • an antigen binding domain against PSCA is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Morgenroth et al., Prostate 67(10):1121-1131 (2007) (scFv 7F5); Nejatollahi et al., J of Oncology 2013(2013), article ID 839831 (scFv C5-II); and US Pat Publication No. 20090311181.
  • CDRs antigen binding portion
  • an antigen binding domain against VEGFR 2 is an antigen binding portion, e.g., 10 CDRs, of an antibody described in, e.g., Chinnasamy et al., J Clin Invest 120(11):3953-3968 (2010).
  • an antigen binding domain against LewisY is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Kelly et al., Cancer Biother Radiopharm 23(4):411-423 (2008) (hu3S193 Ab (scFvs)); Dolezal et al., Protein Engineering 16(1):47-56 (2003) (NC10 scFv).
  • an antigen binding domain against CD24 is an antigen binding portion, e.g., 15 CDRs, of an antibody described in, e.g., Maliar et al., Gastroenterology 143(5):1375-1384 (2012).
  • an antigen binding domain against PDGFR-beta is an antigen binding portion, e.g., CDRs, of an antibody Abcam ab32570.
  • an antigen binding domain against SSEA-4 is an antigen binding portion, e.g., CDRs, of antibody MC813 (Cell Signaling), or other commercially available antibodies.
  • an antigen binding domain against CD20 is an antigen binding portion, e.g., CDRs, of the antibody Rituximab, Ofatumumab, Ocrelizumab, Veltuzumab, or GA101.
  • an antigen binding domain against Folate receptor alpha is an antigen binding portion, e.g., CDRs, of the antibody IMGN853, or an antibody described in US20120009181; US4851332, LK26: US 5952484.
  • an antigen binding portion e.g., CDRs, of the antibody IMGN853, or an antibody described in US20120009181; US4851332, LK26: US 5952484.
  • an antigen binding domain against ERBB2 is an antigen binding portion, e.g., CDRs, of the antibody trastuzumab, or pertuzumab.
  • an antigen binding domain against MUC1 is an antigen binding portion, e.g., CDRs, of the antibody SAR566658.
  • the antigen binding domain against EGFR is antigen binding portion, e.g., 30 CDRs, of the antibody cetuximab, panitumumab, zalutumumab, nimotuzumab, or matuzumab.
  • an antigen binding domain against NCAM is an antigen binding portion, e.g., CDRs, of the antibody clone 2-2B: MAB5324 (EMD Millipore).
  • an antigen binding domain against Ephrin B2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Abengozar et al., Blood 119(19):4565-4576 (2012).
  • an antigen binding domain against IGF-I receptor is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 8344112 B2; EP2322550 A1; WO 2006/138315, or PCT/US2006/022995.
  • an antigen binding domain against CAIX is an antigen binding portion, e.g., CDRs, of the antibody clone 303123 (R&D Systems).
  • an antigen binding domain against LMP2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 7,410,640, or US20050129701.
  • an antigen binding domain against gp100 is an antigen binding portion, e.g., CDRs, of the antibody HMB45, NKIbetaB, or an antibody described in WO2013165940, or US20130295007
  • an antigen binding domain against tyrosinase is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 5843674; or US19950504048.
  • an antigen binding domain against EphA2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Yu et al., Mol Ther 22(1):102-111 (2014).
  • an antigen binding domain against GD3 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 7253263; US 8,207,308; US 20120276046; EP1013761 A3; 20120276046; WO2005035577; or US 6437098.
  • an antigen binding domain against fucosyl GM1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US20100297138; or WO2007/067992.
  • an antigen binding domain against sLe is an antigen binding portion, e.g., CDRs, of the antibody G193 (for lewis Y), see Scott AM et al, Cancer Res 60: 3254-61 (2000), also as described in Neeson et al, J Immunol May 2013190 (Meeting Abstract Supplement) 177.10.
  • an antigen binding domain against GM3 is an antigen binding portion, e.g., CDRs, of the antibody CA 2523449 (mAb 14F7).
  • an antigen binding domain against HMWMAA is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Kmiecik et al., Oncoimmunology 3(1):e27185 (2014) (PMID: 24575382) (mAb9.2.27); US 6528481; WO2010033866; or US 20140004124.
  • an antigen binding portion e.g., CDRs
  • an antigen binding domain against o-acetyl-GD2 is an antigen binding portion, e.g., CDRs, of the antibody 8B6.
  • an antigen binding domain against TEM1/CD248 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Marty et al., Cancer Lett 235(2):298-308 (2006); Zhao et al., J Immunol Methods 363(2):221-232 (2011).
  • an antigen binding domain against CLDN6 is an antigen binding portion, e.g., CDRs, of the antibody IMAB027 (Ganymed Pharmaceuticals), see e.g., clinicaltrial.gov/show/NCT02054351.
  • an antigen binding domain against TSHR is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 8,603,466; US 8,501,415; or US 8,309,693.
  • an antigen binding domain against GPRC5D is an antigen binding portion, e.g., CDRs, of the antibody FAB6300A (R&D Systems); or LS-A4180 (Lifespan Biosciences).
  • an antigen binding domain against CD97 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., US 6,846,911;de Groot et al., J Immunol 183(6):4127-4134 (2009); or an antibody from R&D:MAB3734.
  • an antigen binding domain against ALK is an antigen binding portion, e.g., 5 CDRs, of an antibody described in, e.g., Mino-Kenudson et al., Clin Cancer Res 16(5):1561-1571 (2010).
  • an antigen binding domain against polysialic acid is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Nagae et al., J Biol Chem 288(47):33784-33796 (2013).
  • an antigen binding domain against PLAC1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Ghods et al., Biotechnol Appl Biochem 2013 doi:10.1002/bab.1177. 10
  • an antigen binding domain against GloboH is an antigen binding portion of the antibody VK9; or an antibody described in, e.g., Kudryashov V et al, Glycoconj J.15(3):243-9 ( 1998), Lou et al., Proc Natl Acad Sci USA 111(7):2482-2487 (2014) ; MBr1: Bremer E-G et al. J Biol Chem 259:14773–14777 (1984).
  • an antigen binding domain against NY-BR-1 is an antigen binding portion,15 e.g., CDRs of an antibody described in, e.g., Jager et al., Appl Immunohistochem Mol Morphol 15(1):77- 83 (2007).
  • an antigen binding domain against WT-1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Dao et al., Sci Transl Med 5(176):176ra33 (2013); or WO2012/135854.
  • an antigen binding domain against MAGE-A1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Willemsen et al., J Immunol 174(12):7853-7858 (2005) (TCR- like scFv).
  • an antigen binding domain against sperm protein 17 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Song et al., Target Oncol 2013 Aug 14 (PMID: 25 23943313); Song et al., Med Oncol 29(4):2923-2931 (2012).
  • an antigen binding domain against Tie 2 is an antigen binding portion, e.g., CDRs, of the antibody AB33 (Cell Signaling Technology).
  • an antigen binding domain against MAD-CT-2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., PMID: 2450952; US 7635753.
  • an antigen binding domain against Fos-related antigen 1 is an antigen binding portion, e.g., CDRs, of the antibody 12F9 (Novus Biologicals).
  • an antigen binding domain against MelanA/MART1 is an antigen binding portion, e.g., CDRs, of an antibody described in, EP2514766 A2; or US 7,749,719.
  • an antigen binding domain against sarcoma translocation breakpoints is an35 antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Luo et al, EMBO Mol. Med.4(6):453- 461 (2012).
  • an antigen binding domain against TRP-2 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Wang et al, J Exp Med.184(6):2207-16 (1996).
  • an antigen binding domain against CYP1B1 is an antigen binding portion, e.g., CDRs, of an antibody described in, e.g., Maecker et al, Blood 102 (9): 3287-3294 (2003).
  • an antigen binding domain against RAGE-1 is an antigen binding portion, e.g., CDRs, of the antibody MAB5328 (EMD Millipore).
  • an antigen binding domain against human telomerase reverse transcriptase is an antigen binding portion, e.g., CDRs, of the antibody cat no: LS-B95-100 (Lifespan Biosciences)
  • an antigen binding domain against intestinal carboxyl esterase is an antigen 10 binding portion, e.g., CDRs, of the antibody 4F12: cat no: LS-B6190-50 (Lifespan Biosciences).
  • an antigen binding domain against mut hsp70-2 is an antigen binding portion, e.g., CDRs, of the antibody Lifespan Biosciences: monoclonal: cat no: LS-C133261-100 (Lifespan Biosciences).
  • an antigen binding domain against CD79a is an antigen binding portion, e.g., 15 CDRs, of the antibody Anti-CD79a antibody [HM47/A9] (ab3121), available from Abcam; antibody CD79A Antibody #3351 available from Cell Signaling Technology; or antibody HPA017748 - Anti- CD79A antibody produced in rabbit, available from Sigma Aldrich.
  • an antigen binding domain against CD79b is an antigen binding portion, e.g., CDRs, of the antibody polatuzumab vedotin, anti-CD79b described in Dornan et al.,“Therapeutic potential 20 of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma” Blood.2009 Sep 24;114(13):2721-9. doi: 10.1182/blood-2009-02-205500.
  • CDRs antigen binding portion
  • an antigen binding domain against CD72 is an antigen binding portion, e.g., CDRs, of the antibody J3-109 described in Myers, and Uckun,“An anti-CD72 immunotoxin against therapy-refractory B-lineage acute lymphoblastic leukemia.” Leuk Lymphoma.1995 Jun;18(1-2):119-22, or anti-CD72 (10D6.8.1, mIgG1) described in Polson et al.,“Antibody-Drug Conjugates for the Treatment of Non–Hodgkin's Lymphoma: Target and Linker-Drug Selection” Cancer Res March 15, 200969; 2358.
  • CDRs antigen binding portion
  • an antigen binding domain against LAIR1 is an antigen binding portion, e.g., CDRs, of the antibody ANT-301 LAIR1 antibody, available from ProSpec; or anti-human CD305 (LAIR1) Antibody, available from BioLegend.
  • an antigen binding portion e.g., CDRs, of the antibody ANT-301 LAIR1 antibody, available from ProSpec; or anti-human CD305 (LAIR1) Antibody, available from BioLegend.
  • an antigen binding domain against FCAR is an antigen binding portion, e.g., CDRs, of the antibody CD89/FCARAntibody (Catalog#10414-H08H), available from Sino Biological Inc. 35
  • an antigen binding domain against LILRA2 is an antigen binding portion, e.g., CDRs, of the antibody LILRA2 monoclonal antibody (M17), clone 3C7, available from Abnova, or Mouse Anti-LILRA2 antibody, Monoclonal (2D7), available from Lifespan Biosciences..
  • an antigen binding domain against CD300LF is an antigen binding portion, e.g., CDRs, of the antibody Mouse Anti-CMRF35-like molecule 1 antibody, Monoclonal[UP-D2], available from BioLegend, or Rat Anti-CMRF35-like molecule 1 antibody, Monoclonal[234903], available from R&D Systems.
  • CDRs antigen binding portion

Abstract

La présente invention concerne un composé représenté par la formule (I) ou un sel pharmaceutiquement acceptable, un hydrate, un solvate, un promédicament, un stéréoisomère ou un tautomère de celui-ci, formule dans laquelle R1, R2, Rx, X1, X2, X3, n, nl et q sont tels que définis dans la description, et des procédés de fabrication et d'utilisation de ceux-ci.
PCT/IB2020/051206 2019-02-15 2020-02-13 Dérivés de 3-(1-oxoisoindoline-2-yl)pipéridine-2,6-dione substitués et leurs utilisations WO2020165834A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EA202192029A EA202192029A1 (ru) 2019-02-15 2020-02-13 Замещенные производные 3-(1-оксоизоиндолин-2-ил)пиперидин-2,6-диона и варианты их применения
CA3123519A CA3123519A1 (fr) 2019-02-15 2020-02-13 Derives de 3-(1-oxoisoindoline-2-yl)piperidine-2,6-dione substitues et leurs utilisations
US17/430,478 US20220144798A1 (en) 2019-02-15 2020-02-13 Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
KR1020217028320A KR20210129672A (ko) 2019-02-15 2020-02-13 치환된 3-(1-옥소이소인돌린-2-일)피페리딘-2,6-디온 유도체 및 이의 용도
MX2021009764A MX2021009764A (es) 2019-02-15 2020-02-13 Derivados de 3-(1-oxoisoindolin-2-il)piperidina-2,6-diona sustituidos y usos de los mismos.
CN202080009260.9A CN113329792A (zh) 2019-02-15 2020-02-13 取代的3-(1-氧代异吲哚啉-2-基)哌啶-2,6-二酮衍生物及其用途
JP2021547485A JP2022520448A (ja) 2019-02-15 2020-02-13 置換3-(1-オキソイソインドリン-2-イル)ピペリジン-2,6-ジオン誘導体及びその使用
AU2020222346A AU2020222346B2 (en) 2019-02-15 2020-02-13 Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
EP20708627.3A EP3924055B1 (fr) 2019-02-15 2020-02-13 Dérivés de 3-(1-oxoisoindoline-2-yl)pipéridine-2,6-dione substitués et leurs utilisations

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962806142P 2019-02-15 2019-02-15
US62/806,142 2019-02-15

Publications (1)

Publication Number Publication Date
WO2020165834A1 true WO2020165834A1 (fr) 2020-08-20

Family

ID=69740421

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2020/051206 WO2020165834A1 (fr) 2019-02-15 2020-02-13 Dérivés de 3-(1-oxoisoindoline-2-yl)pipéridine-2,6-dione substitués et leurs utilisations

Country Status (10)

Country Link
US (1) US20220144798A1 (fr)
EP (1) EP3924055B1 (fr)
JP (1) JP2022520448A (fr)
KR (1) KR20210129672A (fr)
CN (1) CN113329792A (fr)
AU (1) AU2020222346B2 (fr)
CA (1) CA3123519A1 (fr)
EA (1) EA202192029A1 (fr)
MX (1) MX2021009764A (fr)
WO (1) WO2020165834A1 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112574094A (zh) * 2020-12-14 2021-03-30 成都大学 吲哚酮衍生物及其制药用途
WO2022216573A1 (fr) * 2021-04-05 2022-10-13 Bristol-Myers Squibb Company Composés d'oxoisoindoline à substitution pyridinyle pour le traitement du cancer
WO2022216644A1 (fr) * 2021-04-06 2022-10-13 Bristol-Myers Squibb Company Composés oxoisoindoline substitués par pyridinyle
CN115252606A (zh) * 2022-09-29 2022-11-01 北京市神经外科研究所 化合物在制备用于***的药物中的应用
WO2023183540A1 (fr) * 2022-03-25 2023-09-28 Regents Of The University Of Michigan Dégradeurs d'ikzf2 et leurs utilisations
WO2023201012A1 (fr) * 2022-04-15 2023-10-19 Regents Of The University Of Michigan Agents de dégradation d'ikzf2 et leurs utilisations
US11878968B2 (en) 2021-07-09 2024-01-23 Plexium, Inc. Aryl compounds and pharmaceutical compositions that modulate IKZF2
US11897862B2 (en) 2022-03-17 2024-02-13 Gilead Sciences, Inc. IKAROS zinc finger family degraders and uses thereof
WO2024073475A1 (fr) * 2022-09-27 2024-04-04 Oncopia Therapeutics, Inc. D/B/A Proteovant Therapeutics, Inc. Ligands de cereblon et utilisations associées

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116640122A (zh) * 2022-02-16 2023-08-25 苏州国匡医药科技有限公司 Ikzf2降解剂及包含其的药物组合物和用途

Citations (359)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
US4261989A (en) 1979-02-19 1981-04-14 Kaken Chemical Co. Ltd. Geldanamycin derivatives and antitumor drug
US4433059A (en) 1981-09-08 1984-02-21 Ortho Diagnostic Systems Inc. Double antibody conjugate
US4444878A (en) 1981-12-21 1984-04-24 Boston Biomedical Research Institute, Inc. Bispecific antibody determinants
US4818541A (en) 1987-08-19 1989-04-04 Schering Corporation Transdermal delivery of enantiomers of phenylpropanolamine
US4851332A (en) 1985-04-01 1989-07-25 Sloan-Kettering Institute For Cancer Research Choriocarcinoma monoclonal antibodies and antibody panels
EP0346087A2 (fr) 1988-06-09 1989-12-13 Snow Brand Milk Products Co., Ltd. Anticorps hybride et procédé pour sa production
WO1991003493A1 (fr) 1989-08-29 1991-03-21 The University Of Southampton CONJUGUES F(ab)3 ou F(ab)4 bi ou trispécifiques
US5114946A (en) 1987-06-12 1992-05-19 American Cyanamid Company Transdermal delivery of pharmaceuticals
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
WO1993023537A1 (fr) 1992-05-08 1993-11-25 Creative Biomolecules Analogues de proteines polyvalents chimeres et procedes d'utilisation
US5273743A (en) 1990-03-09 1993-12-28 Hybritech Incorporated Trifunctional antibody-like compounds as a combined diagnostic and therapeutic agent
WO1994009131A1 (fr) 1992-10-15 1994-04-28 Scotgen Limited Proteine de liaison specifique recombinee
WO1994012625A2 (fr) 1992-11-23 1994-06-09 Zeneca Limited Domaines variables de liaison de ligands (v-min) comprenant une region d'encadrement presentant une permutation cyclique de la structure centrale en baril
WO1995009917A1 (fr) 1993-10-07 1995-04-13 The Regents Of The University Of California Anticorps bispecifiques et tetravalents, obtenus par genie genetique
US5534254A (en) 1992-02-06 1996-07-09 Chiron Corporation Biosynthetic binding proteins for immuno-targeting
WO1996037621A2 (fr) 1995-05-23 1996-11-28 Morphosys Gesellschaft Für Proteinoptimierung Mbh Proteines multimeres
US5582996A (en) 1990-12-04 1996-12-10 The Wistar Institute Of Anatomy & Biology Bifunctional antibodies and method of preparing same
US5591828A (en) 1989-06-22 1997-01-07 Behringwerke Aktiengesellschaft Bispecific and oligospecific mono-and oligovalent receptors, the preparation and use thereof
EP0754230A1 (fr) 1994-04-04 1997-01-22 Genentech, Inc. Anticorps agonistes diriges contre le recepteur flk2/flt3 et leurs utilisations
US5635602A (en) 1993-08-13 1997-06-03 The Regents Of The University Of California Design and synthesis of bispecific DNA-antibody conjugates
US5637481A (en) 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
WO1997025068A2 (fr) 1996-01-05 1997-07-17 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Antigene de mesothelium, procedes et kits de ciblage de celui-ci
EP0805871A1 (fr) 1995-01-18 1997-11-12 Roche Diagnostics GmbH Anticorps anti-cd30 prevenant le clivage proteolytique et la liberation de l'antigene cd30 membranaire
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5777084A (en) 1996-03-07 1998-07-07 Eberhard-Karls-Universitat Tubingen Antibody BV10A4H2 specific for human FLT3/FLK2 receptor and mybridoma
US5786464A (en) 1994-09-19 1998-07-28 The General Hospital Corporation Overexpression of mammalian and viral proteins
US5837242A (en) 1992-12-04 1998-11-17 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US5837821A (en) 1992-11-04 1998-11-17 City Of Hope Antibody construct
US5844094A (en) 1992-09-25 1998-12-01 Commonwealth Scientific And Industrial Research Organization Target binding polypeptide
US5843674A (en) 1993-11-16 1998-12-01 Pola Chemical Industries Inc. Anti-human tyrosinase monoclonal antibody
US5864019A (en) 1990-06-11 1999-01-26 Celltech Limited Multivalent antigen-binding proteins
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US5910573A (en) 1992-01-23 1999-06-08 Merck Patent Gesellschaft Mit Beschrankter Haftung Monomeric and dimeric antibody-fragment fusion proteins
WO1999028471A2 (fr) 1997-12-01 1999-06-10 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services ANTICORPS, NOTAMMENT DES MOLECULES Fv, IMMUNOCONJUGUES PRESENTANT UNE GRANDE AFFINITE DE LIAISON POUR LA MESOTHELINE ET PROCEDES D'UTILISATION CORRESPONDANTS
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
US5952484A (en) 1994-03-08 1999-09-14 Sloan-Kettering Cancer Center Recombinant human anti-LK26 antibodies
US5959083A (en) 1991-06-03 1999-09-28 Behringwerke Aktiengellschaft Tetravalent bispecific receptors, the preparation and use thereof
US5989830A (en) 1995-10-16 1999-11-23 Unilever Patent Holdings Bv Bifunctional or bivalent antibody fragment analogue
WO1999064460A1 (fr) 1998-06-10 1999-12-16 Celltech Therapeutics Limited Fragments d'anticorps bivalents
US6005079A (en) 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
WO2000006605A2 (fr) 1998-07-28 2000-02-10 Micromet Ag Heterominicorps
WO2000035436A2 (fr) 1998-12-16 2000-06-22 Warner-Lambert Company Traitement de l'arthrite a l'aide d'inhibiteurs de la mek
EP1013761A2 (fr) 1991-09-18 2000-06-28 Kyowa Hakko Kogyo Co., Ltd. Anticorps chimérique humanisé dirigé contre le ganglioside GD3
US6114148A (en) 1996-09-20 2000-09-05 The General Hospital Corporation High level expression of proteins
WO2001012812A2 (fr) 1999-08-17 2001-02-22 Biogen, Inc. Recepteur de baff (bcma) et agent immunoregulateur
US6239259B1 (en) 1996-04-04 2001-05-29 Unilever Patent Holdings B.V. Multivalent and multispecific antigen-binding protein
WO2001038490A2 (fr) 1999-11-29 2001-05-31 The Trustees Of Columbia University In The City Of New York ISOLEMENT DE CINQ NOUVEAUX GENES CODANT POUR DES NOUVEAUX RECEPTEURS Fc DE TYPE MELANOME INTERVENANT DANS LA PATHOGENESE DU LYMPHOME MALIN ET DU MELANOME
WO2001051056A1 (fr) 2000-01-10 2001-07-19 Astrazeneca Ab Preparation de fulvestrant
WO2001066139A1 (fr) 2000-03-06 2001-09-13 University Of Kentucky Research Foundation Procedes de degradation de cellules hematologiques embryonnaires cancereuses et composes associes
US6294353B1 (en) 1994-10-20 2001-09-25 Morphosys Ag Targeted hetero-association of recombinant proteins to multi-functional complexes
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
US20020004587A1 (en) 2000-04-11 2002-01-10 Genentech, Inc. Multivalent antibodies and uses therefor
WO2002006213A2 (fr) 2000-07-19 2002-01-24 Warner-Lambert Company Esters oxygenes d'acides 4-iodophenylamino benzhydroxamiques
WO2002016436A2 (fr) 2000-08-22 2002-02-28 Novartis Ag ANTICORPS DE LA IL-1β HUMAINE
US20020076406A1 (en) 2000-07-25 2002-06-20 Leung Shui-On Multivalent target binding protein
US20020103345A1 (en) 2000-05-24 2002-08-01 Zhenping Zhu Bispecific immunoglobulin-like antigen binding proteins and method of production
WO2002066470A1 (fr) 2001-01-12 2002-08-29 Amgen Inc. Derives d'alkylamine substitues et methodes d'utilisation
WO2002072635A2 (fr) 2001-03-13 2002-09-19 University College London Elements de liaison specifiques
US6476198B1 (en) 1993-07-13 2002-11-05 The Scripps Research Institute Multispecific and multivalent antigen-binding polypeptide molecules
US6511663B1 (en) 1991-06-11 2003-01-28 Celltech R&D Limited Tri- and tetra-valent monospecific antigen-binding proteins
US6528481B1 (en) 1999-02-16 2003-03-04 The Burnam Institute NG2/HM proteoglycan-binding peptides that home to angiogenic vasculature and related methods
WO2003062401A2 (fr) 2002-01-22 2003-07-31 Corixa Corporation Compositions et methodes de detection, de diagnostic et de traitement des malignites hematologiques
WO2003064383A2 (fr) 2002-02-01 2003-08-07 Ariad Gene Therapeutics, Inc. Composés contenant du phosphore et utilisations associées
WO2003076424A1 (fr) 2002-03-08 2003-09-18 Eisai Co. Ltd. Composes macrocycliques utiles comme produits pharmaceutiques
WO2003077914A1 (fr) 2002-03-13 2003-09-25 Array Biopharma, Inc Utilisation de derives de benzimidazole alkyles n3 en tant qu'inhibiteurs de mek
US20030207346A1 (en) 1997-05-02 2003-11-06 William R. Arathoon Method for making multispecific antibodies having heteromultimeric and common components
US20030211078A1 (en) 2001-12-07 2003-11-13 Heavner George A. Pseudo-antibody constructs
US6670453B2 (en) 1997-10-27 2003-12-30 Unilever Patent Holdings B.V. Multivalent antigen-binding proteins
WO2004005284A1 (fr) 2002-07-09 2004-01-15 Astrazeneca Ab 3-cyanoquinoleines substituees utilisees comme inhibiteurs de mek
WO2004007529A2 (fr) 2002-07-15 2004-01-22 The Trustees Of Princeton University Composes qui se lient a iap
US6743896B2 (en) 1997-04-30 2004-06-01 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
WO2004045532A2 (fr) 2002-11-15 2004-06-03 Chiron Corporation Procedes de prevention et de traitement de metastase cancereuse et de perte osseuse liee a la metastase cancereuse
US6770622B2 (en) 2001-06-08 2004-08-03 Gary A. Jarvis N-terminally truncated galectin-3 for use in treating cancer
US6780996B2 (en) 2002-04-30 2004-08-24 Wyeth Holdings Corporation Process for the preparation of 7-substituted-3 quinolinecarbonitriles
WO2004081051A1 (fr) 2003-03-12 2004-09-23 The University Of Birmingham Anticorps specifiques
WO2004087758A2 (fr) 2003-03-26 2004-10-14 Neopharm, Inc. Anticorps du recepteur alpha 2 il 13 et procedes d'utilisation
US6809185B1 (en) 1998-01-23 2004-10-26 Vlaams Interuniversitair Instituut Voor Biotechnologie Multipurpose antibody derivatives
US20040220388A1 (en) 2000-06-30 2004-11-04 Nico Mertens Novel heterodimeric fusion proteins
US20040219643A1 (en) 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US20040242847A1 (en) 2000-10-20 2004-12-02 Naoshi Fukushima Degraded agonist antibody
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
US20050003403A1 (en) 2003-04-22 2005-01-06 Rossi Edmund A. Polyvalent protein complex
US20050004352A1 (en) 1998-04-09 2005-01-06 Roland Kontermann Single-chain multiple antigen-binding molecule, its preparation and use
US6846911B2 (en) 1996-10-25 2005-01-25 The United States Of America As Represented By The Department Of Health And Human Services Methods and compositions for inhibiting inflammation and angiogenesis comprising a mammalian CD97 α subunit
WO2005014652A1 (fr) 2003-08-05 2005-02-17 Morphotek, Inc. Molecule a surface cellulaire variante liee au cancer
US20050069552A1 (en) 2003-07-28 2005-03-31 Bleck Gregory T. Fusion antibodies
WO2005028443A2 (fr) 2003-09-15 2005-03-31 Wyeth A Corporation Of The State Of Delaware, Usa Inhibiteurs de l'enzyme de la proteine tyrosine kinase
US20050079170A1 (en) 2001-09-14 2005-04-14 Fabrice Le Gall Dimeric and multimeric antigen binding structure
WO2005035577A1 (fr) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Compositions d'anticorps se liant specifiquement au ganglioside gd3
US20050100543A1 (en) 2003-07-01 2005-05-12 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
US20050129701A1 (en) 2001-12-04 2005-06-16 Marasco Wayne A. Antibody to latent membrane proteins and uses thereof
US20050136051A1 (en) 2003-12-22 2005-06-23 Bernard Scallon Methods for generating multimeric molecules
US20050136049A1 (en) 2001-01-17 2005-06-23 Ledbetter Jeffrey A. Binding constructs and methods for use thereof
US20050163782A1 (en) 2003-06-27 2005-07-28 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
WO2005068503A2 (fr) 2004-01-07 2005-07-28 Chiron Corporation Anticorps monoclonal specifique du m-csf et ses utilisations
WO2005069888A2 (fr) 2004-01-16 2005-08-04 The Regents Of The University Of Michigan Peptidomimetiques de smac et utilisations associees
WO2005069894A2 (fr) 2004-01-16 2005-08-04 The Regents Of The University Of Michigan Mimetiques de smac contraints de maniere conformationnelle et utilisations associees
US20050175606A1 (en) 2001-04-11 2005-08-11 Hua-Liang Huang Cyclic single-chain trispecific antibody
WO2005094818A1 (fr) 2004-03-23 2005-10-13 Genentech, Inc. Inhibiteurs azabicyclo-octane de l'iap
WO2005097791A1 (fr) 2004-04-07 2005-10-20 Novartis Ag Inhibiteurs d'iap
US20050266425A1 (en) 2003-12-31 2005-12-01 Vaccinex, Inc. Methods for producing and identifying multispecific antibodies
WO2005117986A2 (fr) 2004-06-01 2005-12-15 Genentech, Inc. Conjugues de medicaments anticorps et procedes correspondants
WO2005121142A1 (fr) 2004-06-11 2005-12-22 Japan Tobacco Inc. Dérivés de 5-amino-2,4,7-trioxo-3,4,7,8-tétrahydro-2h-pyrido’2,3-d! pyrimidine et composés apparentés pour le traitement du cancer
US20060014700A1 (en) 2004-07-02 2006-01-19 Genentech, Inc. Inhibitors of IAP
WO2006010118A2 (fr) 2004-07-09 2006-01-26 The Regents Of The University Of Michigan Mimetiques de smac contraints par conformation et utilisations de ceux-ci
US20060025347A1 (en) 2004-07-15 2006-02-02 Condon Stephen M IAP binding compounds
WO2006017295A2 (fr) 2004-07-12 2006-02-16 Idun Pharmaceuticals, Inc. Analogues de tetrapeptide
WO2006020258A2 (fr) 2004-07-17 2006-02-23 Imclone Systems Incorporated Nouveau anticorps bispecifique tetravalent
WO2006039238A2 (fr) 2004-09-30 2006-04-13 The Goverment Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Anticorps de irta2 et méthodes d'utilisation
US20060083747A1 (en) 2002-12-27 2006-04-20 Domantis Limited Fc fusion
US20060120960A1 (en) 2004-01-30 2006-06-08 Sergey Deyev Multivalent complexes, their production and method of use
WO2006069063A1 (fr) 2004-12-20 2006-06-29 Genentech, Inc. Inhibiteurs des iap derives de la pyrrolidine
WO2006076691A2 (fr) 2005-01-12 2006-07-20 Medarex, Inc. Anticorps 2 associes a la translocation de recepteur immunitaire (irta-2) et leurs utilisations
US7090843B1 (en) 2000-11-28 2006-08-15 Seattle Genetics, Inc. Recombinant anti-CD30 antibodies and uses thereof
WO2006086469A2 (fr) 2005-02-08 2006-08-17 Genzyme Corporation Anticorps anti-tgf-beta
US20060204493A1 (en) 2004-09-02 2006-09-14 Genentech, Inc. Heteromultimeric molecules
WO2006099141A2 (fr) 2005-03-10 2006-09-21 Morphotek, Inc. Anticorps diriges contre la mesotheline
WO2006105021A2 (fr) 2005-03-25 2006-10-05 Tolerrx, Inc. Molecules de liaison gitr et leurs utilisations
WO2006106905A1 (fr) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha Procede pour la production de polypeptide au moyen de la regulation d’un ensemble
US7129330B1 (en) 1998-05-05 2006-10-31 Deutsches Krebsforschungszentrum Stiftung Des Offentlichen Rechts Multivalent antibody constructs
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
US20060263367A1 (en) 2005-05-23 2006-11-23 Fey Georg H Bispecific antibody devoid of Fc region and method of treatment using same
WO2006125481A1 (fr) 2005-05-27 2006-11-30 Universitätsklinikum Freiburg Anticorps monoclonaux et fragments d'anticorps a chaine unique contre antigene de membrane specifique a la prostate ayant pour origine la surface cellulaire
WO2006138315A2 (fr) 2005-06-15 2006-12-28 Schering Corporation Formulation d'anticorps stable
US20070004909A1 (en) 2005-04-15 2007-01-04 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2007004606A1 (fr) 2005-07-04 2007-01-11 Nikon Vision Co., Ltd. Appareil de mesure de distance
WO2007004415A1 (fr) 2005-07-01 2007-01-11 Murata Manufacturing Co., Ltd. Substrat céramique à couches multiples, procédé pour le fabriquer et feuille verte composite pour la fabrication dudit substrat
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
WO2007014011A2 (fr) 2005-07-21 2007-02-01 Ardea Biosciences, Inc. Inhibiteurs n-(arylamino)-sulfonamide de mek
US7183076B2 (en) 1997-05-02 2007-02-27 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
WO2007024715A2 (fr) 2005-08-19 2007-03-01 Abbott Laboratories Immunoglobuline a deux domaines variables et utilisations de celle-ci
WO2007044887A2 (fr) 2005-10-11 2007-04-19 Transtarget, Inc. Procede de production d'une population homogene d'anticorps bispecifiques tetravalents
US20070087381A1 (en) 2002-04-15 2007-04-19 Tetsuo Kojima Methods for constructing scdb libraries
US20070128150A1 (en) 2003-12-23 2007-06-07 Norman Timothy J Branched molecular scaffolds for linking polymer residues to biologically active moieties
WO2007067992A2 (fr) 2005-12-08 2007-06-14 Medarex, Inc. Anticorps monoclonaux humains se liant au fucosyl-gm1, et procedes d'utilisation de l'anti-fucosyl-gm1
US20070141049A1 (en) 2005-08-26 2007-06-21 Reinhard Bredehorst Bivalent IgY antibody constructs for diagnostic and therapeutic applications
US20070154901A1 (en) 1997-06-11 2007-07-05 Protein Engineering Technology Aps Trimerising module
WO2007084342A2 (fr) 2006-01-13 2007-07-26 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services, National Institutes Of Health Il-15 et il-15r-alpha améliorées aux fins d'expression dans des cellules mammaliennes
US7253263B1 (en) 1999-09-30 2007-08-07 Kyowa Hakko Kogyo Co., Ltd. Complementarity determining region-grafted antibody against ganglioside GD3 and derivative of antibody against ganglioside GD3
WO2007095338A2 (fr) 2006-02-15 2007-08-23 Imclone Systems Incorporated Formulation d'anticorps
WO2007110205A2 (fr) 2006-03-24 2007-10-04 Merck Patent Gmbh Domaines de proteine heterodimerique d'ingenierie
WO2007121484A2 (fr) 2006-04-19 2007-10-25 Novartis Ag Composés à base de benzoxazole et de benzothiazole 6-0 substitués et procédés d'inhibition de signalisation csf-1r
US20070274985A1 (en) 2006-05-26 2007-11-29 Stefan Dubel Antibody
WO2007137760A2 (fr) 2006-05-25 2007-12-06 Bayer Schering Pharma Aktiengesellschaft Complexes moléculaires dimères
WO2008024725A1 (fr) 2006-08-21 2008-02-28 Genentech, Inc. Composés aza-benzofuranyle et leurs procédés d'utilisation
US20080050370A1 (en) 2006-03-17 2008-02-28 Scott Glaser Stabilized polypeptide compositions
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008061741A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Dérivés de 5-sulfanylméthyl-[1,2,4]triazol[1, 5-a]pyrimidin-7-ol utilisés comme antagonistes de cxcr2
WO2008062026A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Derives de 5-sulfanylmethyl-pyrazolo[1,5-a]pyrimidin-7-ol utilises en tant qu'antagonistes du cxcr2
WO2008061740A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Pyrimidines et leur utilisation comme antagonistes du récepteur cxcr2
US20080152645A1 (en) 2006-08-18 2008-06-26 Armagen Technologies, Inc. Genetically Encoded Multifunctional Compositions Bidrectionally Transported Between Peripheral Blood and the CNS
US7410640B2 (en) 1998-07-22 2008-08-12 Vanderbilt University GBS toxin receptor antibodies
US20080241884A1 (en) 2003-10-08 2008-10-02 Kenya Shitara Fused Protein Composition
WO2008119353A1 (fr) 2007-03-29 2008-10-09 Genmab A/S Anticorps bispécifiques et procédés de production de ceux-ci
US20080254512A1 (en) 2006-11-02 2008-10-16 Capon Daniel J Hybrid immunoglobulins with moving parts
US20080260738A1 (en) 2007-04-18 2008-10-23 Moore Margaret D Single chain fc, methods of making and methods of treatment
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
WO2008134679A1 (fr) 2007-04-30 2008-11-06 Genentech, Inc. Inhibiteurs de iap
WO2008143794A1 (fr) 2007-05-11 2008-11-27 Altor Bioscience Corporation Molécules de fusion et variantes de il-15
WO2008146911A1 (fr) 2007-06-01 2008-12-04 Sapporo Medical University Anticorps dirigé contre il13ra2 et agent de diagnostic/thérapie comprenant l'anticorps
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2009021754A2 (fr) 2007-08-15 2009-02-19 Bayer Schering Pharma Aktiengesellschaft Anticorps monospécifiques et multispécifiques, et procédés d'utilisation
WO2009036082A2 (fr) 2007-09-12 2009-03-19 Genentech, Inc. Combinaisons de composés inhibiteurs des phosphoinositide 3-kinases et agents chimiothérapeutiques, et leurs procédés d'utilisation
WO2009045957A1 (fr) 2007-10-01 2009-04-09 Medarex, Inc. Anticorps humains qui se lient à la mésothéline, et utilisations de ceux-ci
WO2009044273A2 (fr) 2007-10-05 2009-04-09 Immutep Utilisation d'une protéine lag-3 recombinée ou de dérivés de celle-ci pour produire une réponse immunitaire des monocytes
US7521056B2 (en) 2005-04-06 2009-04-21 Ibc Pharmaceuticals, Inc. Stably tethered structures of defined compositions with multiple functions or binding specificities
WO2009055730A1 (fr) 2007-10-25 2009-04-30 Genentech, Inc. Procédé de préparation de composés de thiénopyrimidine
US7527787B2 (en) 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
US20090130106A1 (en) 2005-11-29 2009-05-21 The University Of Sydney Demibodies: dimerization-activated therapeutic agents
WO2009068204A1 (fr) 2007-11-26 2009-06-04 Bayer Schering Pharma Aktiengesellschaft Anticorps anti-mésothéline et leurs utilisations
WO2009068630A1 (fr) 2007-11-27 2009-06-04 Ablynx N.V. Constructions d'immunoglobuline
US20090148905A1 (en) 2007-11-30 2009-06-11 Claire Ashman Antigen-binding constructs
US20090155275A1 (en) 2007-07-31 2009-06-18 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US20090162360A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US20090175851A1 (en) 2007-12-21 2009-07-09 Christian Klein Bivalent, bispecific antibodies
WO2009085983A1 (fr) 2007-12-19 2009-07-09 Genentech, Inc. 5-anilinoimidazopyridines et procédés d'utilisation
US20090175867A1 (en) 2006-06-12 2009-07-09 Trubion Pharmaceuticals, Inc. Single-Chain Multivalent Binding Proteins with Effector Function
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
WO2009106539A1 (fr) 2008-02-26 2009-09-03 Novartis Ag Composés hétérocycliques comme inhibiteurs de cxcr2
US20090232811A1 (en) 2007-12-21 2009-09-17 Christian Klein Bivalent, bispecific antibodies
US20090234105A1 (en) 2006-03-24 2009-09-17 The Regents Of The University Of California Construction of a Multivalent SCFV Through Alkyne-Azide 1,3-Dipolar Cycloaddition
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US20090252742A1 (en) 2007-04-13 2009-10-08 Ivan Bergstein IL3Ralpha antibody conjugates and uses thereof
US20090263392A1 (en) 2006-03-31 2009-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US7612114B2 (en) 2002-12-26 2009-11-03 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20090274649A1 (en) 2002-03-01 2009-11-05 Immunomedics, Inc. Bispecific Antibody Point Mutations for Enhancing Rate of Clearance
US20090297529A1 (en) 2008-05-30 2009-12-03 Yiwen Li Anti-flt3 antibodies
US20090304657A1 (en) 2006-05-03 2009-12-10 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Chimeric t cell receptors and related materials and methods of use
US20090304718A1 (en) 2006-01-05 2009-12-10 Guenther Adolf Antibody Molecules Specific for Fibroblast Activation Protein and Immunoconjugates Containing Them
US20090311181A1 (en) 2006-03-20 2009-12-17 The Regents Of The University Of California Engineered Anti-Prostate Stem Cell Antigen (PSCA) Antibodies for Cancer Targeting
US7635753B2 (en) 2007-02-19 2009-12-22 Wisconsin Alumni Research Foundation Prostate cancer and melanoma antigens
WO2009155386A1 (fr) 2008-06-20 2009-12-23 Abbott Laboratories Procédé pour préparer le promoteur d'apoptose abt-263
WO2009156737A1 (fr) 2008-06-25 2009-12-30 Vernalis (R&D) Limited Dérivés de triazalo [4, 5-d] pyramidine et leur utilisation comme antagonistes des récepteurs de la purine
WO2010015613A1 (fr) 2008-08-04 2010-02-11 Novartis Ag Composés organiques
WO2010019570A2 (fr) 2008-08-11 2010-02-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
WO2010020675A1 (fr) 2008-08-22 2010-02-25 Novartis Ag Composés de pyrrolopyrimidine et leurs utilisations
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010033866A2 (fr) 2008-09-19 2010-03-25 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Anticorps monoclonaux de cspg4 utilises dans le diagnostic et le traitement du carcinome mammaire de type basal
WO2010063802A1 (fr) 2008-12-05 2010-06-10 Novartis Ag Cyclobutène-1,2-diones 3,4-disubstituées en tant qu'antagonistes de récepteur cxcr2
US20100150910A1 (en) 2006-10-10 2010-06-17 Universite De Nantes Use of monoclonal antibodies specific to the o-acetylated form of gd2 ganglioside for treatment of certain cancers
US7749719B2 (en) 1994-04-22 2010-07-06 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US7767675B2 (en) 2006-11-22 2010-08-03 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
WO2010114940A1 (fr) 2009-04-01 2010-10-07 Genentech, Inc. Anticorps anti-fcrh5 et immunoconjugués et procédés d'utilisation
WO2010120561A1 (fr) 2009-04-01 2010-10-21 Genentech, Inc. Anticorps et immunoconjugués anti-fcrh5 et procédés d'utilisation
WO2010126066A1 (fr) 2009-04-27 2010-11-04 協和発酵キリン株式会社 Anticorps anti-il-3rα destiné à être utilisé dans le traitement d'hématomes
WO2010129304A2 (fr) 2009-04-27 2010-11-11 Oncomed Pharmaceuticals, Inc. Procédé de fabrication de molécules hétéromultimères
WO2011028683A1 (fr) 2009-09-03 2011-03-10 Schering Corporation Anticorps anti-gitr
US7915391B2 (en) 2006-04-24 2011-03-29 Amgen Inc. Humanized c-Kit antibody
WO2011049677A1 (fr) 2009-09-02 2011-04-28 Irm Llc Composés et compositions utilisés en tant que modulateurs de l'activité tlr
EP2322550A1 (fr) 2004-12-22 2011-05-18 Amgen, Inc Compositions conprenant anti-IGF-1R anticorps et méthodes pour leur production
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
WO2011076786A1 (fr) 2009-12-22 2011-06-30 Novartis Ag Isoquinolinones et quinazolinones substituées
WO2011076922A1 (fr) 2009-12-23 2011-06-30 Synimmune Gmbh Anticorps anti-flt3 et leurs méthodes d'emploi
WO2011095625A1 (fr) 2010-02-05 2011-08-11 Heptares Therapeutics Limited Dérivés de 1,2,4-triazine-4-amine
WO2011131746A2 (fr) 2010-04-20 2011-10-27 Genmab A/S Protéines contenant des anticorps fc hétérodimères et leurs procédés de production
US20110268656A1 (en) 2009-12-02 2011-11-03 David Ho J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
WO2011156518A2 (fr) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Modulateur du récepteur oestrogénique et utilisation de ces derniers
US8080650B2 (en) 2003-06-27 2011-12-20 Diadexus, Inc. Pro104 antibody compositions and methods of use
WO2011159847A2 (fr) 2010-06-15 2011-12-22 The Regents Of The University Of California Conjugués du fragment d'anticorps fv à chaîne unique dirigé contre le récepteur orphelin 1 analogue au récepteur à la tyrosine kinase (ror1) et leurs procédés d'utilisation
WO2011160119A2 (fr) 2010-06-19 2011-12-22 Memorial Sloan-Kettering Cancer Center Anticorps contre gd2
WO2011159769A2 (fr) 2010-06-17 2011-12-22 Aragon Pharmaceuticals, Inc. Modulateurs de récepteur d'œstrogène d'indane et utilisations de ceux-ci
US20120009181A1 (en) 2010-02-24 2012-01-12 Ab Olga Folate Receptor 1 Antibodies and Immunoconjugates and Uses Thereof
US8114845B2 (en) 2008-08-25 2012-02-14 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US20120039906A1 (en) 2009-02-09 2012-02-16 INSER (Institut National de la Recherche Medicale) PD-1 Antibodies and PD-L1 Antibodies and Uses Thereof
WO2012033885A1 (fr) 2010-09-08 2012-03-15 Baylor College Of Medicine Immunothérapie des cancers utilisant des lymphocytes t génétiquement modifiés, spécifiques de gd2
WO2012037410A2 (fr) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Modulateurs des récepteurs des oestrogènes et leurs utilisations
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
WO2012062713A1 (fr) 2010-11-08 2012-05-18 Novartis Ag Polypeptides se liant aux récepteurs de chimiokines
WO2012079000A1 (fr) 2010-12-09 2012-06-14 The Trustees Of The University Of Pennsylvania Utilisation de lymphocytes t modifiés par un récepteur chimérique d'antigènes chimérique pour traiter le cancer
US8207228B2 (en) 2004-10-04 2012-06-26 Regents Of The University Of Minnesota Calixarene-based peptide conformation mimetics, methods of use, and methods of making
US8207308B2 (en) 2007-02-16 2012-06-26 Sloan-Kettering Institute For Cancer Research Anti ganglioside GD3 antibodies and uses thereof
US8236780B2 (en) 2006-05-16 2012-08-07 Galectin Therapeutics Galactose—pronged polysaccharides in a formulation for antifibrotic therapies
US8263746B2 (en) 2004-02-06 2012-09-11 Morphosys Ag Anti-CD38 human antibodies and uses thereof
WO2012135854A2 (fr) 2011-04-01 2012-10-04 Memorial Sloan-Kettering Cancer Center Anticorps contre des peptides cytosoliques
EP2514766A2 (fr) 2007-03-29 2012-10-24 Technion Research & Development Foundation Ltd. Anticorps, procédés et kits pour diagnostiquer et traiter un mélanome
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
US8309693B2 (en) 2001-08-23 2012-11-13 Rsr Limited Epitope regions of a thyrotrophin (TSH) receptor, uses thereof and antibodies thereto
US20120288506A1 (en) 2011-05-12 2012-11-15 Imclone Llc C-kit antibodies and uses thereof
WO2012167143A1 (fr) 2011-06-03 2012-12-06 Xoma Technology Ltd. Anticorps spécifiques du tgf bêta
WO2012163805A1 (fr) 2011-05-27 2012-12-06 Glaxo Group Limited Protéines de liaison à bcma (cd269/tnfrsf17)
WO2012175222A1 (fr) 2011-06-24 2012-12-27 Cytune Immunocytokines à base d'il-15 et domaine sushi d'il-15rα
US8344112B2 (en) 2007-07-31 2013-01-01 Merck Sharp & Dohme Limited IGF-1R specific antibodies useful in the detection and diagnosis of cellular proliferative disorders
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US8362211B2 (en) 2010-12-30 2013-01-29 Takeda Pharmaceutical Company Limited Anti-CD38 antibodies
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013030803A1 (fr) 2011-09-02 2013-03-07 Novartis Ag Sel de choline d'un composé anti-inflammatoire à base de cyclobutènedione substitué
US8399645B2 (en) 2003-11-05 2013-03-19 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
WO2013040371A2 (fr) 2011-09-16 2013-03-21 Baylor College Of Medicine Ciblage du microenvironnement tumoral au moyen de cellules nkt modifiées
WO2013040557A2 (fr) 2011-09-16 2013-03-21 The Trustees Of The University Of Pennsylvania Lymphocytes t à arn modifié pour le traitement du cancer
WO2013061273A1 (fr) 2011-10-25 2013-05-02 Massimo Dominici Cellule effectrice modifiée (ou récepteur chimérique) pour traiter la néoplasie exprimant le disialoganglioside gd2
WO2013063419A2 (fr) 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania Récepteur immunitaire chimérique spécifique complètement humain, anti-mésothéline pour un ciblage redirigé de cellules exprimant la mésothéline
WO2013060867A2 (fr) 2011-10-27 2013-05-02 Genmab A/S Production de protéines hétérodimères
US8440798B2 (en) 2006-10-04 2013-05-14 Københavns Universitet Generation of a cancer-specific immune response toward MUC1 and cancer specific MUC1 antibodies
WO2013074916A1 (fr) 2011-11-18 2013-05-23 Board Of Regents, The University Of Texas System Lymphocytes t car+ génétiquement modifiés pour éliminer l'expression du récepteur des lymphocytes t et/ou le système hla
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
WO2013085552A1 (fr) 2011-12-08 2013-06-13 Cleveland Clinic Foundation Fixation de la voûte glénoïde
WO2013111105A1 (fr) 2012-01-26 2013-08-01 Novartis Ag Composés imidazopyrrolidinone
US8501415B2 (en) 2002-11-26 2013-08-06 B.R.A.H.M.S. Gmbh Identification of TSH receptor autoantibodies using affinity-purified antibodies
WO2013123061A1 (fr) 2012-02-13 2013-08-22 Seattle Children's Hospital D/B/A Seattle Children's Research Institute Récepteurs d'antigène chimères bispécifiques et utilisations thérapeutiques de ceux-ci
WO2013126712A1 (fr) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Topicompositions et procédés pour produire une population de lymphocytes t tenaces utiles dans le traitement du cancer
WO2013142034A1 (fr) 2012-03-23 2013-09-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Récepteurs d'antigène chimérique anti-mésothéline
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
US8552156B2 (en) 2010-06-11 2013-10-08 Kyowa Hakko Kirin Co., Ltd Anti-TIM-3 antibody
WO2013165940A1 (fr) 2012-05-01 2013-11-07 Genentech, Inc. Anticorps anti-pmel17 et immunoconjugués
WO2013168108A2 (fr) 2012-05-09 2013-11-14 Novartis Ag Polypeptides de liaison de récepteur de chimiokine
WO2013173820A2 (fr) 2012-05-18 2013-11-21 Scott & White Healthcare Immunofusion bispécifique (ifb) de scfv
WO2013179174A1 (fr) 2012-05-29 2013-12-05 Koninklijke Philips N.V. Système d'éclairage
US8603466B2 (en) 2006-03-29 2013-12-10 King's College London Agonist antibodies against TSHR
WO2013192294A1 (fr) 2012-06-20 2013-12-27 Boston 3T Biotechnologies, Inc. Thérapies cellulaires pour le traitement et la prévention de cancers et d'autres troubles du système immunitaire
WO2014022758A1 (fr) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Anticorps de liaison double à agent unique anti-pd-l1 et pd-l2 et procédés d'utilisation
WO2014031687A1 (fr) 2012-08-20 2014-02-27 Jensen, Michael Procédé et compositions pour l'immunothérapie cellulaire
US20140086932A1 (en) 2012-09-17 2014-03-27 Peter G. Traber Method for enhancing specific immunotherapies in cancer treatment
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
WO2014066527A2 (fr) 2012-10-24 2014-05-01 Admune Therapeutics Llc Formes d'il-15r alpha, cellules exprimant des formes d'il-15r alpha, et utilisations thérapeutiques d'il-15r alpha et de complexes il-15/il-15r alpha
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2014089335A2 (fr) 2012-12-07 2014-06-12 Amgen Inc. Protéines de liaison à l'antigène bcma
WO2014100079A1 (fr) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Anticorps qui se lient au ligand 1 de la mort programmée humaine (pd-l1)
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
US8796284B2 (en) 2010-03-31 2014-08-05 Palobiofarma, S.L. 4-aminopyrimidine derivatives and their as as adenosine A2a receptor antagonists
WO2014122144A1 (fr) 2013-02-05 2014-08-14 Engmab Ag Anticorps bispécifiques anti-cd3ɛ et bcma
WO2014130657A1 (fr) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Traitement du cancer au moyen d'un récepteur d'antigènes chimériques anti-egfrviii humanisés
WO2014130635A1 (fr) 2013-02-20 2014-08-28 Novartis Ag Ciblage efficace de la leucémie primaire humaine au moyen de lymphocytes t génétiquement modifiés des récepteurs d'antigènes chimériques anti-cd123
WO2014130310A1 (fr) 2013-02-19 2014-08-28 Novartis Ag Dérivés de benzothiophène et compositions correspondantes en tant qu'agents de dégradation sélectifs des récepteurs des œstrogènes
WO2014138819A1 (fr) 2013-03-14 2014-09-18 Csl Limited Agents qui neutralisent la signalisation par il-3 et leurs utilisations
WO2014140180A1 (fr) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Protéines de liaison anti-lag-3
WO2014138805A1 (fr) 2013-03-14 2014-09-18 Csl Limited Agents anti-il-3r alpha et leurs utilisations
WO2014144622A2 (fr) 2013-03-15 2014-09-18 Stephen Forman Lymphocytes t redirigés par des récepteurs d'antigènes chimériques spécifiques de cd123 et leurs procédés d'utilisation
WO2014140248A1 (fr) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Molécules liantes pour bcma et cd3
US8841418B2 (en) 2011-07-01 2014-09-23 Cellerant Therapeutics, Inc. Antibodies that specifically bind to TIM3
WO2014153270A1 (fr) 2013-03-16 2014-09-25 Novartis Ag Traitement du cancer à l'aide d'un récepteur d'antigène chimérique anti-cd19 humanisé
WO2014165707A2 (fr) 2013-04-03 2014-10-09 Memorial Sloan-Kettering Cancer Center Génération efficace de lymphocytes t ciblant une tumeur dérivés de cellules souches pluripotentes
WO2014179664A2 (fr) 2013-05-02 2014-11-06 Anaptysbio, Inc. Anticorps dirigés contre la protéine de mort programmée 1 (pd-1)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2014210064A1 (fr) 2013-06-24 2014-12-31 Genentech, Inc. Anticorps anti-fcrh5
WO2014209804A1 (fr) 2013-06-24 2014-12-31 Biomed Valley Discoveries, Inc. Anticorps bispécifiques
US8927697B2 (en) 2008-09-12 2015-01-06 Isis Innovation Limited PD-1 specific antibodies and uses thereof
WO2015026684A1 (fr) 2013-08-20 2015-02-26 Merck Sharp & Dohme Corp. Modulation d'immunité tumorale
WO2015031667A2 (fr) 2013-08-30 2015-03-05 Amgen Inc. Protéines de liaison à l'antigène gitr
US8993731B2 (en) 2010-03-11 2015-03-31 Ucb Biopharma Sprl PD-1 antibody
WO2015061668A1 (fr) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux anti-pd-l1 et fragments de ceux-ci
US9029393B2 (en) 2009-01-26 2015-05-12 Kaldi Pharma, Sas Adenosine receptor ligands and uses thereof
WO2015081158A1 (fr) 2013-11-26 2015-06-04 Bristol-Myers Squibb Company Procédé de traitement du vih par perturbation de la signalisation pd-1/pd-l1
WO2015085847A1 (fr) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Anticorps anti-pd-1, son fragment de liaison à l'antigène, et son application médicale
WO2015090230A1 (fr) 2013-12-19 2015-06-25 Novartis Ag Récepteurs antigéniques chimériques de la mésothéline humaine et leurs utilisations
WO2015109124A2 (fr) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Agents immunomodulateurs
WO2015112805A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains dirigés contre pd-l1
US20150210769A1 (en) 2014-01-24 2015-07-30 Novartis Ag Antibody molecules to pd-1 and uses thereof
WO2015112800A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains se liant à pd-1
WO2015116539A1 (fr) 2014-01-28 2015-08-06 Bristol-Myers Squibb Company Anticorps anti-lag-3 pour traiter des hémopathies malignes
US20150218274A1 (en) 2014-01-31 2015-08-06 Novartis Ag Antibody molecules to tim-3 and uses thereof
US9133197B2 (en) 2009-03-20 2015-09-15 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Oxidated derivatives of triazolylpurines useful as ligands of the adenosine A2A receptor and their use as medicaments
US20150259420A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015136017A1 (fr) 2014-03-13 2015-09-17 F. Hoffmann-La Roche Ag Méthodes et compositions pour moduler des mutants du récepteur des oestrogènes
WO2015142675A2 (fr) 2014-03-15 2015-09-24 Novartis Ag Traitement du cancer au moyen d'un récepteur antigénique chimérique
US9163087B2 (en) 2010-06-18 2015-10-20 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against TIM-3 and PD-1 for immunotherapy in chronic immune conditions
US9175082B2 (en) 2012-05-31 2015-11-03 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
WO2015184099A1 (fr) 2014-05-28 2015-12-03 4-Antibody Ag Anticorps anti-gitr et leurs procédés d'utilisation
WO2015181342A1 (fr) 2014-05-29 2015-12-03 Spring Bioscience Corporation Anticorps dirigés contre pd-l1 et leurs utilisations
WO2015195163A1 (fr) 2014-06-20 2015-12-23 R-Pharm Overseas, Inc. Anticorps totalement humain anti-pd-l1
WO2015200119A1 (fr) 2014-06-26 2015-12-30 Macrogenics, Inc. Dianticorps liés par covalence, présentant une immunoréactivité avec pd-1 et lag-3 et leurs procédés d'utilisation
US9228016B2 (en) 2014-06-06 2016-01-05 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
WO2016000619A1 (fr) 2014-07-03 2016-01-07 Beigene, Ltd. Anticorps anti-pd-l1 et leur utilisation comme agents thérapeutiques et diagnostiques
US9244059B2 (en) 2007-04-30 2016-01-26 Immutep Parc Club Orsay Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2016014535A1 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer au moyen d'un récepteur d'antigènes chimériques cll-1
WO2016014789A2 (fr) 2014-07-24 2016-01-28 Bluebird Bio, Inc. Récepteurs de l'antigène chimérique bcma
WO2016014565A2 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer au moyen d'un récepteur d'antigène chimérique anti-bcma humanisé
WO2016014576A1 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer à l'aide du récepteur antigénique chimérique anti-cd33
WO2016028896A1 (fr) 2014-08-19 2016-02-25 Novartis Ag Récepteur d'antigène chimérique anti-cd123 (car) utilisé dans le traitement du cancer
WO2016028672A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps et fragments de fixation à l'antigène anti-lag3
WO2016054638A1 (fr) 2014-10-03 2016-04-07 Dana-Farber Cancer Institute, Inc. Anticorps dirigés contre le récepteur du facteur de nécrose tumorale induit par glucocorticoïdes (gitr) et leurs procédés d'utilisation
WO2016057846A1 (fr) 2014-10-08 2016-04-14 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer
US20160108123A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016071448A1 (fr) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Anticorps anti-tim3 et procédés d'utilisation
US20160129108A1 (en) 2014-11-11 2016-05-12 Medimmune Limited Therapeutic combinations comprising anti-cd73 antibodies and uses thereof
WO2016092419A1 (fr) 2014-12-09 2016-06-16 Rinat Neuroscience Corp. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
WO2016111947A2 (fr) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Anticorps inhibiteurs d'interactions de tim-3:lilrb2 et leurs utilisations
WO2016144803A2 (fr) 2015-03-06 2016-09-15 Sorrento Therapeutics, Inc. Anticorps thérapeutiques se liant à tim3
WO2016161270A1 (fr) 2015-04-01 2016-10-06 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de cellule t et protéine 3 de mucine (tim-3)
US9505839B2 (en) 2012-07-02 2016-11-29 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
WO2016196792A1 (fr) 2015-06-03 2016-12-08 Bristol-Myers Squibb Company Anticorps anti-gitr pour le diagnostic du cancer
US20170015758A1 (en) 2014-01-21 2017-01-19 Medimmune, Llc Compositions And Methods For Modulating And Redirecting Immune Responses
WO2017015623A2 (fr) 2015-07-23 2017-01-26 Inhibrx Lp Protéines hybrides multivalentes et multispécifiques se liant à gitr
WO2017025918A1 (fr) 2015-08-11 2017-02-16 Novartis Ag 5-bromo -2,6-di- (1h-pyrazol-1-yl)pyrimidin-4-amine pour utilisation dans le traitement du cancer
WO2017027645A1 (fr) 2015-08-13 2017-02-16 Merck Sharp & Dohme Corp. Composés di-nucléotidiques cycliques en tant qu'agonistes de sting
WO2017025610A1 (fr) 2015-08-12 2017-02-16 Medimmune Limited Protéines de fusion gitrl et leurs utilisations
US20180099940A1 (en) * 2016-10-11 2018-04-12 Arvinas, Inc. Compounds and methods for the targeted degradation of androgen receptor
WO2018102725A1 (fr) * 2016-12-01 2018-06-07 Arvinas, Inc. Dérivés de tétrahydronaphtalène et de tétrahydroisoquinoléine en tant qu'agents de dégradation des récepteurs des œstrogènes
WO2018140809A1 (fr) * 2017-01-26 2018-08-02 Arvinas, Inc. Modulateurs du récepteur des œstrogènes de protéolyse et procédés d'utilisation associés
WO2019038717A1 (fr) 2017-08-23 2019-02-28 Novartis Ag Dérivés de 3-(1-oxoisoindolin-2-yl)pipéridine-2,6-dione et leurs utilisations
WO2019079569A1 (fr) * 2017-10-18 2019-04-25 Novartis Ag Compositions et méthodes pour la dégradation sélective d'une protéine

Patent Citations (405)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2779780A (en) 1955-03-01 1957-01-29 Du Pont 1, 4-diamino-2, 3-dicyano-1, 4-bis (substituted mercapto) butadienes and their preparation
US4261989A (en) 1979-02-19 1981-04-14 Kaken Chemical Co. Ltd. Geldanamycin derivatives and antitumor drug
US4433059A (en) 1981-09-08 1984-02-21 Ortho Diagnostic Systems Inc. Double antibody conjugate
US4444878A (en) 1981-12-21 1984-04-24 Boston Biomedical Research Institute, Inc. Bispecific antibody determinants
US4851332A (en) 1985-04-01 1989-07-25 Sloan-Kettering Institute For Cancer Research Choriocarcinoma monoclonal antibodies and antibody panels
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US5114946A (en) 1987-06-12 1992-05-19 American Cyanamid Company Transdermal delivery of pharmaceuticals
US4818541A (en) 1987-08-19 1989-04-04 Schering Corporation Transdermal delivery of enantiomers of phenylpropanolamine
EP0346087A2 (fr) 1988-06-09 1989-12-13 Snow Brand Milk Products Co., Ltd. Anticorps hybride et procédé pour sa production
US5591828A (en) 1989-06-22 1997-01-07 Behringwerke Aktiengesellschaft Bispecific and oligospecific mono-and oligovalent receptors, the preparation and use thereof
WO1991003493A1 (fr) 1989-08-29 1991-03-21 The University Of Southampton CONJUGUES F(ab)3 ou F(ab)4 bi ou trispécifiques
US5273743A (en) 1990-03-09 1993-12-28 Hybritech Incorporated Trifunctional antibody-like compounds as a combined diagnostic and therapeutic agent
US5864019A (en) 1990-06-11 1999-01-26 Celltech Limited Multivalent antigen-binding proteins
US5582996A (en) 1990-12-04 1996-12-10 The Wistar Institute Of Anatomy & Biology Bifunctional antibodies and method of preparing same
US5959083A (en) 1991-06-03 1999-09-28 Behringwerke Aktiengellschaft Tetravalent bispecific receptors, the preparation and use thereof
US6511663B1 (en) 1991-06-11 2003-01-28 Celltech R&D Limited Tri- and tetra-valent monospecific antigen-binding proteins
EP1013761A2 (fr) 1991-09-18 2000-06-28 Kyowa Hakko Kogyo Co., Ltd. Anticorps chimérique humanisé dirigé contre le ganglioside GD3
US6437098B1 (en) 1991-09-18 2002-08-20 Kyowa Hakko Kogyo Co., Ltd. Human chimeric antibody specific for the ganglioside GD3
US5932448A (en) 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
US5910573A (en) 1992-01-23 1999-06-08 Merck Patent Gesellschaft Mit Beschrankter Haftung Monomeric and dimeric antibody-fragment fusion proteins
US5534254A (en) 1992-02-06 1996-07-09 Chiron Corporation Biosynthetic binding proteins for immuno-targeting
WO1993023537A1 (fr) 1992-05-08 1993-11-25 Creative Biomolecules Analogues de proteines polyvalents chimeres et procedes d'utilisation
US6005079A (en) 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
US5844094A (en) 1992-09-25 1998-12-01 Commonwealth Scientific And Industrial Research Organization Target binding polypeptide
WO1994009131A1 (fr) 1992-10-15 1994-04-28 Scotgen Limited Proteine de liaison specifique recombinee
US5262564A (en) 1992-10-30 1993-11-16 Octamer, Inc. Sulfinic acid adducts of organo nitroso compounds useful as retroviral inactivating agents anti-retroviral agents and anti-tumor agents
US5837821A (en) 1992-11-04 1998-11-17 City Of Hope Antibody construct
WO1994012625A2 (fr) 1992-11-23 1994-06-09 Zeneca Limited Domaines variables de liaison de ligands (v-min) comprenant une region d'encadrement presentant une permutation cyclique de la structure centrale en baril
US5837242A (en) 1992-12-04 1998-11-17 Medical Research Council Multivalent and multispecific binding proteins, their manufacture and use
US5637481A (en) 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US6476198B1 (en) 1993-07-13 2002-11-05 The Scripps Research Institute Multispecific and multivalent antigen-binding polypeptide molecules
US5635602A (en) 1993-08-13 1997-06-03 The Regents Of The University Of California Design and synthesis of bispecific DNA-antibody conjugates
WO1995009917A1 (fr) 1993-10-07 1995-04-13 The Regents Of The University Of California Anticorps bispecifiques et tetravalents, obtenus par genie genetique
US5843674A (en) 1993-11-16 1998-12-01 Pola Chemical Industries Inc. Anti-human tyrosinase monoclonal antibody
US5952484A (en) 1994-03-08 1999-09-14 Sloan-Kettering Cancer Center Recombinant human anti-LK26 antibodies
EP0754230A1 (fr) 1994-04-04 1997-01-22 Genentech, Inc. Anticorps agonistes diriges contre le recepteur flk2/flt3 et leurs utilisations
US7749719B2 (en) 1994-04-22 2010-07-06 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
US5786464A (en) 1994-09-19 1998-07-28 The General Hospital Corporation Overexpression of mammalian and viral proteins
US5786464C1 (en) 1994-09-19 2012-04-24 Gen Hospital Corp Overexpression of mammalian and viral proteins
US6294353B1 (en) 1994-10-20 2001-09-25 Morphosys Ag Targeted hetero-association of recombinant proteins to multi-functional complexes
EP0805871A1 (fr) 1995-01-18 1997-11-12 Roche Diagnostics GmbH Anticorps anti-cd30 prevenant le clivage proteolytique et la liberation de l'antigene cd30 membranaire
US20070014794A1 (en) 1995-03-01 2007-01-18 Genentech, Inc. Method for making heteromultimeric polypeptides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
WO1996037621A2 (fr) 1995-05-23 1996-11-28 Morphosys Gesellschaft Für Proteinoptimierung Mbh Proteines multimeres
US5989830A (en) 1995-10-16 1999-11-23 Unilever Patent Holdings Bv Bifunctional or bivalent antibody fragment analogue
WO1997025068A2 (fr) 1996-01-05 1997-07-17 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Antigene de mesothelium, procedes et kits de ciblage de celui-ci
US5777084A (en) 1996-03-07 1998-07-07 Eberhard-Karls-Universitat Tubingen Antibody BV10A4H2 specific for human FLT3/FLK2 receptor and mybridoma
US6239259B1 (en) 1996-04-04 2001-05-29 Unilever Patent Holdings B.V. Multivalent and multispecific antigen-binding protein
US6114148A (en) 1996-09-20 2000-09-05 The General Hospital Corporation High level expression of proteins
US6114148C1 (en) 1996-09-20 2012-05-01 Gen Hospital Corp High level expression of proteins
US6846911B2 (en) 1996-10-25 2005-01-25 The United States Of America As Represented By The Department Of Health And Human Services Methods and compositions for inhibiting inflammation and angiogenesis comprising a mammalian CD97 α subunit
US6743896B2 (en) 1997-04-30 2004-06-01 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
US20030207346A1 (en) 1997-05-02 2003-11-06 William R. Arathoon Method for making multispecific antibodies having heteromultimeric and common components
US7183076B2 (en) 1997-05-02 2007-02-27 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20070154901A1 (en) 1997-06-11 2007-07-05 Protein Engineering Technology Aps Trimerising module
US6670453B2 (en) 1997-10-27 2003-12-30 Unilever Patent Holdings B.V. Multivalent antigen-binding proteins
WO1999028471A2 (fr) 1997-12-01 1999-06-10 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services ANTICORPS, NOTAMMENT DES MOLECULES Fv, IMMUNOCONJUGUES PRESENTANT UNE GRANDE AFFINITE DE LIAISON POUR LA MESOTHELINE ET PROCEDES D'UTILISATION CORRESPONDANTS
US6809185B1 (en) 1998-01-23 2004-10-26 Vlaams Interuniversitair Instituut Voor Biotechnologie Multipurpose antibody derivatives
US20050004352A1 (en) 1998-04-09 2005-01-06 Roland Kontermann Single-chain multiple antigen-binding molecule, its preparation and use
US7129330B1 (en) 1998-05-05 2006-10-31 Deutsches Krebsforschungszentrum Stiftung Des Offentlichen Rechts Multivalent antibody constructs
WO1999064460A1 (fr) 1998-06-10 1999-12-16 Celltech Therapeutics Limited Fragments d'anticorps bivalents
US7410640B2 (en) 1998-07-22 2008-08-12 Vanderbilt University GBS toxin receptor antibodies
WO2000006605A2 (fr) 1998-07-28 2000-02-10 Micromet Ag Heterominicorps
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
WO2000035436A2 (fr) 1998-12-16 2000-06-22 Warner-Lambert Company Traitement de l'arthrite a l'aide d'inhibiteurs de la mek
US6528481B1 (en) 1999-02-16 2003-03-04 The Burnam Institute NG2/HM proteoglycan-binding peptides that home to angiogenic vasculature and related methods
WO2001012812A2 (fr) 1999-08-17 2001-02-22 Biogen, Inc. Recepteur de baff (bcma) et agent immunoregulateur
US7253263B1 (en) 1999-09-30 2007-08-07 Kyowa Hakko Kogyo Co., Ltd. Complementarity determining region-grafted antibody against ganglioside GD3 and derivative of antibody against ganglioside GD3
WO2001038490A2 (fr) 1999-11-29 2001-05-31 The Trustees Of Columbia University In The City Of New York ISOLEMENT DE CINQ NOUVEAUX GENES CODANT POUR DES NOUVEAUX RECEPTEURS Fc DE TYPE MELANOME INTERVENANT DANS LA PATHOGENESE DU LYMPHOME MALIN ET DU MELANOME
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
WO2001051056A1 (fr) 2000-01-10 2001-07-19 Astrazeneca Ab Preparation de fulvestrant
WO2001066139A1 (fr) 2000-03-06 2001-09-13 University Of Kentucky Research Foundation Procedes de degradation de cellules hematologiques embryonnaires cancereuses et composes associes
US20020004587A1 (en) 2000-04-11 2002-01-10 Genentech, Inc. Multivalent antibodies and uses therefor
US20020103345A1 (en) 2000-05-24 2002-08-01 Zhenping Zhu Bispecific immunoglobulin-like antigen binding proteins and method of production
US20040220388A1 (en) 2000-06-30 2004-11-04 Nico Mertens Novel heterodimeric fusion proteins
WO2002006213A2 (fr) 2000-07-19 2002-01-24 Warner-Lambert Company Esters oxygenes d'acides 4-iodophenylamino benzhydroxamiques
US20020076406A1 (en) 2000-07-25 2002-06-20 Leung Shui-On Multivalent target binding protein
US7446175B2 (en) 2000-08-22 2008-11-04 Novartis Ag Antibodies to human IL-1β
WO2002016436A2 (fr) 2000-08-22 2002-02-28 Novartis Ag ANTICORPS DE LA IL-1β HUMAINE
EP1313769A2 (fr) 2000-08-22 2003-05-28 Novartis AG Anticorps de la il-1 beta humaine
US20040242847A1 (en) 2000-10-20 2004-12-02 Naoshi Fukushima Degraded agonist antibody
US7090843B1 (en) 2000-11-28 2006-08-15 Seattle Genetics, Inc. Recombinant anti-CD30 antibodies and uses thereof
WO2002066470A1 (fr) 2001-01-12 2002-08-29 Amgen Inc. Derives d'alkylamine substitues et methodes d'utilisation
US20050136049A1 (en) 2001-01-17 2005-06-23 Ledbetter Jeffrey A. Binding constructs and methods for use thereof
WO2002072635A2 (fr) 2001-03-13 2002-09-19 University College London Elements de liaison specifiques
US20050175606A1 (en) 2001-04-11 2005-08-11 Hua-Liang Huang Cyclic single-chain trispecific antibody
US6770622B2 (en) 2001-06-08 2004-08-03 Gary A. Jarvis N-terminally truncated galectin-3 for use in treating cancer
US20040219643A1 (en) 2001-06-28 2004-11-04 Greg Winter Dual-specific ligand
US6833441B2 (en) 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
US8309693B2 (en) 2001-08-23 2012-11-13 Rsr Limited Epitope regions of a thyrotrophin (TSH) receptor, uses thereof and antibodies thereto
US20050079170A1 (en) 2001-09-14 2005-04-14 Fabrice Le Gall Dimeric and multimeric antigen binding structure
US20050129701A1 (en) 2001-12-04 2005-06-16 Marasco Wayne A. Antibody to latent membrane proteins and uses thereof
US20030211078A1 (en) 2001-12-07 2003-11-13 Heavner George A. Pseudo-antibody constructs
WO2003062401A2 (fr) 2002-01-22 2003-07-31 Corixa Corporation Compositions et methodes de detection, de diagnostic et de traitement des malignites hematologiques
WO2003064383A2 (fr) 2002-02-01 2003-08-07 Ariad Gene Therapeutics, Inc. Composés contenant du phosphore et utilisations associées
US20090274649A1 (en) 2002-03-01 2009-11-05 Immunomedics, Inc. Bispecific Antibody Point Mutations for Enhancing Rate of Clearance
WO2003076424A1 (fr) 2002-03-08 2003-09-18 Eisai Co. Ltd. Composes macrocycliques utiles comme produits pharmaceutiques
WO2003077914A1 (fr) 2002-03-13 2003-09-25 Array Biopharma, Inc Utilisation de derives de benzimidazole alkyles n3 en tant qu'inhibiteurs de mek
US20070087381A1 (en) 2002-04-15 2007-04-19 Tetsuo Kojima Methods for constructing scdb libraries
US6780996B2 (en) 2002-04-30 2004-08-24 Wyeth Holdings Corporation Process for the preparation of 7-substituted-3 quinolinecarbonitriles
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
WO2004005284A1 (fr) 2002-07-09 2004-01-15 Astrazeneca Ab 3-cyanoquinoleines substituees utilisees comme inhibiteurs de mek
WO2004007529A2 (fr) 2002-07-15 2004-01-22 The Trustees Of Princeton University Composes qui se lient a iap
WO2004045532A2 (fr) 2002-11-15 2004-06-03 Chiron Corporation Procedes de prevention et de traitement de metastase cancereuse et de perte osseuse liee a la metastase cancereuse
US8501415B2 (en) 2002-11-26 2013-08-06 B.R.A.H.M.S. Gmbh Identification of TSH receptor autoantibodies using affinity-purified antibodies
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
US20100028330A1 (en) 2002-12-23 2010-02-04 Medimmune Limited Methods of upmodulating adaptive immune response using anti-pd1 antibodies
US7612114B2 (en) 2002-12-26 2009-11-03 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator
US20060083747A1 (en) 2002-12-27 2006-04-20 Domantis Limited Fc fusion
WO2004081051A1 (fr) 2003-03-12 2004-09-23 The University Of Birmingham Anticorps specifiques
WO2004087758A2 (fr) 2003-03-26 2004-10-14 Neopharm, Inc. Anticorps du recepteur alpha 2 il 13 et procedes d'utilisation
US20080171855A1 (en) 2003-04-22 2008-07-17 Ibc Pharmaceuticals, Inc. Polyvalent protein complex
US20050003403A1 (en) 2003-04-22 2005-01-06 Rossi Edmund A. Polyvalent protein complex
US20050163782A1 (en) 2003-06-27 2005-07-28 Biogen Idec Ma Inc. Modified binding molecules comprising connecting peptides
US8080650B2 (en) 2003-06-27 2011-12-20 Diadexus, Inc. Pro104 antibody compositions and methods of use
US20050100543A1 (en) 2003-07-01 2005-05-12 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
US20050069552A1 (en) 2003-07-28 2005-03-31 Bleck Gregory T. Fusion antibodies
WO2005014652A1 (fr) 2003-08-05 2005-02-17 Morphotek, Inc. Molecule a surface cellulaire variante liee au cancer
WO2005028443A2 (fr) 2003-09-15 2005-03-31 Wyeth A Corporation Of The State Of Delaware, Usa Inhibiteurs de l'enzyme de la proteine tyrosine kinase
WO2005035577A1 (fr) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Compositions d'anticorps se liant specifiquement au ganglioside gd3
US20080241884A1 (en) 2003-10-08 2008-10-02 Kenya Shitara Fused Protein Composition
US8399645B2 (en) 2003-11-05 2013-03-19 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US20050136051A1 (en) 2003-12-22 2005-06-23 Bernard Scallon Methods for generating multimeric molecules
US20070128150A1 (en) 2003-12-23 2007-06-07 Norman Timothy J Branched molecular scaffolds for linking polymer residues to biologically active moieties
US20050266425A1 (en) 2003-12-31 2005-12-01 Vaccinex, Inc. Methods for producing and identifying multispecific antibodies
WO2005068503A2 (fr) 2004-01-07 2005-07-28 Chiron Corporation Anticorps monoclonal specifique du m-csf et ses utilisations
US9079956B2 (en) 2004-01-07 2015-07-14 Novartis Vaccines And Diagnostics Inc. M-CSF specific monoclonal antibody and uses thereof
WO2005069888A2 (fr) 2004-01-16 2005-08-04 The Regents Of The University Of Michigan Peptidomimetiques de smac et utilisations associees
WO2005069894A2 (fr) 2004-01-16 2005-08-04 The Regents Of The University Of Michigan Mimetiques de smac contraints de maniere conformationnelle et utilisations associees
US20060120960A1 (en) 2004-01-30 2006-06-08 Sergey Deyev Multivalent complexes, their production and method of use
US8263746B2 (en) 2004-02-06 2012-09-11 Morphosys Ag Anti-CD38 human antibodies and uses thereof
WO2005094818A1 (fr) 2004-03-23 2005-10-13 Genentech, Inc. Inhibiteurs azabicyclo-octane de l'iap
WO2005097791A1 (fr) 2004-04-07 2005-10-20 Novartis Ag Inhibiteurs d'iap
WO2005117986A2 (fr) 2004-06-01 2005-12-15 Genentech, Inc. Conjugues de medicaments anticorps et procedes correspondants
WO2005121142A1 (fr) 2004-06-11 2005-12-22 Japan Tobacco Inc. Dérivés de 5-amino-2,4,7-trioxo-3,4,7,8-tétrahydro-2h-pyrido’2,3-d! pyrimidine et composés apparentés pour le traitement du cancer
US20060014700A1 (en) 2004-07-02 2006-01-19 Genentech, Inc. Inhibitors of IAP
WO2006010118A2 (fr) 2004-07-09 2006-01-26 The Regents Of The University Of Michigan Mimetiques de smac contraints par conformation et utilisations de ceux-ci
WO2006017295A2 (fr) 2004-07-12 2006-02-16 Idun Pharmaceuticals, Inc. Analogues de tetrapeptide
US20060025347A1 (en) 2004-07-15 2006-02-02 Condon Stephen M IAP binding compounds
WO2006020258A2 (fr) 2004-07-17 2006-02-23 Imclone Systems Incorporated Nouveau anticorps bispecifique tetravalent
US20060204493A1 (en) 2004-09-02 2006-09-14 Genentech, Inc. Heteromultimeric molecules
WO2006039238A2 (fr) 2004-09-30 2006-04-13 The Goverment Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Anticorps de irta2 et méthodes d'utilisation
US8207228B2 (en) 2004-10-04 2012-06-26 Regents Of The University Of Minnesota Calixarene-based peptide conformation mimetics, methods of use, and methods of making
WO2006069063A1 (fr) 2004-12-20 2006-06-29 Genentech, Inc. Inhibiteurs des iap derives de la pyrrolidine
EP2322550A1 (fr) 2004-12-22 2011-05-18 Amgen, Inc Compositions conprenant anti-IGF-1R anticorps et méthodes pour leur production
WO2006076691A2 (fr) 2005-01-12 2006-07-20 Medarex, Inc. Anticorps 2 associes a la translocation de recepteur immunitaire (irta-2) et leurs utilisations
US8591901B2 (en) 2005-02-08 2013-11-26 Genzyme Corporation Antibodies to TGF-β
WO2006086469A2 (fr) 2005-02-08 2006-08-17 Genzyme Corporation Anticorps anti-tgf-beta
US8383780B2 (en) 2005-02-08 2013-02-26 Genzyme Corporation Antibodies to TGFβ
WO2006099141A2 (fr) 2005-03-10 2006-09-21 Morphotek, Inc. Anticorps diriges contre la mesotheline
US7812135B2 (en) 2005-03-25 2010-10-12 Tolerrx, Inc. GITR-binding antibodies
US9028823B2 (en) 2005-03-25 2015-05-12 Gitr, Inc. Methods of inducing or enhancing an immune response in a subject by administering agonistic GITR binding antibodies
WO2006105021A2 (fr) 2005-03-25 2006-10-05 Tolerrx, Inc. Molecules de liaison gitr et leurs utilisations
US8388967B2 (en) 2005-03-25 2013-03-05 Gitr, Inc. Methods for inducing or enhancing an immune response by administering agonistic GITR-binding antibodies
WO2006106905A1 (fr) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha Procede pour la production de polypeptide au moyen de la regulation d’un ensemble
US7521056B2 (en) 2005-04-06 2009-04-21 Ibc Pharmaceuticals, Inc. Stably tethered structures of defined compositions with multiple functions or binding specificities
US20070004909A1 (en) 2005-04-15 2007-01-04 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US20060263367A1 (en) 2005-05-23 2006-11-23 Fey Georg H Bispecific antibody devoid of Fc region and method of treatment using same
WO2006125481A1 (fr) 2005-05-27 2006-11-30 Universitätsklinikum Freiburg Anticorps monoclonaux et fragments d'anticorps a chaine unique contre antigene de membrane specifique a la prostate ayant pour origine la surface cellulaire
WO2006138315A2 (fr) 2005-06-15 2006-12-28 Schering Corporation Formulation d'anticorps stable
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
WO2007004415A1 (fr) 2005-07-01 2007-01-11 Murata Manufacturing Co., Ltd. Substrat céramique à couches multiples, procédé pour le fabriquer et feuille verte composite pour la fabrication dudit substrat
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007004606A1 (fr) 2005-07-04 2007-01-11 Nikon Vision Co., Ltd. Appareil de mesure de distance
WO2007014011A2 (fr) 2005-07-21 2007-02-01 Ardea Biosciences, Inc. Inhibiteurs n-(arylamino)-sulfonamide de mek
WO2007024715A2 (fr) 2005-08-19 2007-03-01 Abbott Laboratories Immunoglobuline a deux domaines variables et utilisations de celle-ci
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070141049A1 (en) 2005-08-26 2007-06-21 Reinhard Bredehorst Bivalent IgY antibody constructs for diagnostic and therapeutic applications
WO2007044887A2 (fr) 2005-10-11 2007-04-19 Transtarget, Inc. Procede de production d'une population homogene d'anticorps bispecifiques tetravalents
US7527787B2 (en) 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
US20090130106A1 (en) 2005-11-29 2009-05-21 The University Of Sydney Demibodies: dimerization-activated therapeutic agents
WO2007067992A2 (fr) 2005-12-08 2007-06-14 Medarex, Inc. Anticorps monoclonaux humains se liant au fucosyl-gm1, et procedes d'utilisation de l'anti-fucosyl-gm1
US20100297138A1 (en) 2005-12-08 2010-11-25 Vistica Cynthia A Human monoclonal antibodies to fucosyl-gm1 and methods for using anti-fucosyl-gm1
US20090304718A1 (en) 2006-01-05 2009-12-10 Guenther Adolf Antibody Molecules Specific for Fibroblast Activation Protein and Immunoconjugates Containing Them
WO2007084342A2 (fr) 2006-01-13 2007-07-26 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services, National Institutes Of Health Il-15 et il-15r-alpha améliorées aux fins d'expression dans des cellules mammaliennes
WO2007095338A2 (fr) 2006-02-15 2007-08-23 Imclone Systems Incorporated Formulation d'anticorps
US20080050370A1 (en) 2006-03-17 2008-02-28 Scott Glaser Stabilized polypeptide compositions
US20090311181A1 (en) 2006-03-20 2009-12-17 The Regents Of The University Of California Engineered Anti-Prostate Stem Cell Antigen (PSCA) Antibodies for Cancer Targeting
WO2007110205A2 (fr) 2006-03-24 2007-10-04 Merck Patent Gmbh Domaines de proteine heterodimerique d'ingenierie
US20090234105A1 (en) 2006-03-24 2009-09-17 The Regents Of The University Of California Construction of a Multivalent SCFV Through Alkyne-Azide 1,3-Dipolar Cycloaddition
US8603466B2 (en) 2006-03-29 2013-12-10 King's College London Agonist antibodies against TSHR
US20090263392A1 (en) 2006-03-31 2009-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US7553854B2 (en) 2006-04-19 2009-06-30 Novartis Vaccines And Diagnostics, Inc. 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
WO2007121484A2 (fr) 2006-04-19 2007-10-25 Novartis Ag Composés à base de benzoxazole et de benzothiazole 6-0 substitués et procédés d'inhibition de signalisation csf-1r
US8173689B2 (en) 2006-04-19 2012-05-08 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
US8710048B2 (en) 2006-04-19 2014-04-29 Novartis Ag 6-O-substituted benzoxazole and benzothiazole compounds and methods of inhibiting CSF-1R signaling
US7915391B2 (en) 2006-04-24 2011-03-29 Amgen Inc. Humanized c-Kit antibody
US20090304657A1 (en) 2006-05-03 2009-12-10 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Chimeric t cell receptors and related materials and methods of use
US8236780B2 (en) 2006-05-16 2012-08-07 Galectin Therapeutics Galactose—pronged polysaccharides in a formulation for antifibrotic therapies
WO2007137760A2 (fr) 2006-05-25 2007-12-06 Bayer Schering Pharma Aktiengesellschaft Complexes moléculaires dimères
US20070274985A1 (en) 2006-05-26 2007-11-29 Stefan Dubel Antibody
US20090175867A1 (en) 2006-06-12 2009-07-09 Trubion Pharmaceuticals, Inc. Single-Chain Multivalent Binding Proteins with Effector Function
US20080152645A1 (en) 2006-08-18 2008-06-26 Armagen Technologies, Inc. Genetically Encoded Multifunctional Compositions Bidrectionally Transported Between Peripheral Blood and the CNS
WO2008024725A1 (fr) 2006-08-21 2008-02-28 Genentech, Inc. Composés aza-benzofuranyle et leurs procédés d'utilisation
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
US8440798B2 (en) 2006-10-04 2013-05-14 Københavns Universitet Generation of a cancer-specific immune response toward MUC1 and cancer specific MUC1 antibodies
US20100150910A1 (en) 2006-10-10 2010-06-17 Universite De Nantes Use of monoclonal antibodies specific to the o-acetylated form of gd2 ganglioside for treatment of certain cancers
US20080254512A1 (en) 2006-11-02 2008-10-16 Capon Daniel J Hybrid immunoglobulins with moving parts
US8461330B2 (en) 2006-11-22 2013-06-11 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
US7767675B2 (en) 2006-11-22 2010-08-03 Incyte Corporation Imidazotriazines and imidazopyrimidines as kinase inhibitors
WO2008062026A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Derives de 5-sulfanylmethyl-pyrazolo[1,5-a]pyrimidin-7-ol utilises en tant qu'antagonistes du cxcr2
US20100152205A1 (en) 2006-11-23 2010-06-17 Novartis Ag Cxcr2 inhibitors
WO2008061740A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Pyrimidines et leur utilisation comme antagonistes du récepteur cxcr2
WO2008061741A1 (fr) 2006-11-23 2008-05-29 Novartis Ag Dérivés de 5-sulfanylméthyl-[1,2,4]triazol[1, 5-a]pyrimidin-7-ol utilisés comme antagonistes de cxcr2
US20120276046A1 (en) 2007-02-16 2012-11-01 Sloan-Kettering Institute For Cancer Research Anti ganglioside gd3 antibodies and uses thereof
US8207308B2 (en) 2007-02-16 2012-06-26 Sloan-Kettering Institute For Cancer Research Anti ganglioside GD3 antibodies and uses thereof
US7635753B2 (en) 2007-02-19 2009-12-22 Wisconsin Alumni Research Foundation Prostate cancer and melanoma antigens
WO2008119353A1 (fr) 2007-03-29 2008-10-09 Genmab A/S Anticorps bispécifiques et procédés de production de ceux-ci
EP2514766A2 (fr) 2007-03-29 2012-10-24 Technion Research & Development Foundation Ltd. Anticorps, procédés et kits pour diagnostiquer et traiter un mélanome
US20090252742A1 (en) 2007-04-13 2009-10-08 Ivan Bergstein IL3Ralpha antibody conjugates and uses thereof
US20080260738A1 (en) 2007-04-18 2008-10-23 Moore Margaret D Single chain fc, methods of making and methods of treatment
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
WO2008134679A1 (fr) 2007-04-30 2008-11-06 Genentech, Inc. Inhibiteurs de iap
US9244059B2 (en) 2007-04-30 2016-01-26 Immutep Parc Club Orsay Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2008143794A1 (fr) 2007-05-11 2008-11-27 Altor Bioscience Corporation Molécules de fusion et variantes de il-15
WO2008146911A1 (fr) 2007-06-01 2008-12-04 Sapporo Medical University Anticorps dirigé contre il13ra2 et agent de diagnostic/thérapie comprenant l'anticorps
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US8344112B2 (en) 2007-07-31 2013-01-01 Merck Sharp & Dohme Limited IGF-1R specific antibodies useful in the detection and diagnosis of cellular proliferative disorders
US20090155275A1 (en) 2007-07-31 2009-06-18 Medimmune, Llc Multispecific epitope binding proteins and uses thereof
WO2009021754A2 (fr) 2007-08-15 2009-02-19 Bayer Schering Pharma Aktiengesellschaft Anticorps monospécifiques et multispécifiques, et procédés d'utilisation
WO2009036082A2 (fr) 2007-09-12 2009-03-19 Genentech, Inc. Combinaisons de composés inhibiteurs des phosphoinositide 3-kinases et agents chimiothérapeutiques, et leurs procédés d'utilisation
WO2009045957A1 (fr) 2007-10-01 2009-04-09 Medarex, Inc. Anticorps humains qui se lient à la mésothéline, et utilisations de ceux-ci
WO2009044273A2 (fr) 2007-10-05 2009-04-09 Immutep Utilisation d'une protéine lag-3 recombinée ou de dérivés de celle-ci pour produire une réponse immunitaire des monocytes
WO2009055730A1 (fr) 2007-10-25 2009-04-30 Genentech, Inc. Procédé de préparation de composés de thiénopyrimidine
WO2009068204A1 (fr) 2007-11-26 2009-06-04 Bayer Schering Pharma Aktiengesellschaft Anticorps anti-mésothéline et leurs utilisations
WO2009068630A1 (fr) 2007-11-27 2009-06-04 Ablynx N.V. Constructions d'immunoglobuline
US20090148905A1 (en) 2007-11-30 2009-06-11 Claire Ashman Antigen-binding constructs
WO2009085983A1 (fr) 2007-12-19 2009-07-09 Genentech, Inc. 5-anilinoimidazopyridines et procédés d'utilisation
US20090162360A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
US20090175851A1 (en) 2007-12-21 2009-07-09 Christian Klein Bivalent, bispecific antibodies
US20090232811A1 (en) 2007-12-21 2009-09-17 Christian Klein Bivalent, bispecific antibodies
US20090162359A1 (en) 2007-12-21 2009-06-25 Christian Klein Bivalent, bispecific antibodies
WO2009089004A1 (fr) 2008-01-07 2009-07-16 Amgen Inc. Méthode de fabrication de molécules hétérodimères fc d'anticorps utilisant les effets de conduite électrostatique
WO2009101611A1 (fr) 2008-02-11 2009-08-20 Curetech Ltd. Anticorps monoclonaux pour le traitement de tumeurs
WO2009106539A1 (fr) 2008-02-26 2009-09-03 Novartis Ag Composés hétérocycliques comme inhibiteurs de cxcr2
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
US20090297529A1 (en) 2008-05-30 2009-12-03 Yiwen Li Anti-flt3 antibodies
WO2009155386A1 (fr) 2008-06-20 2009-12-23 Abbott Laboratories Procédé pour préparer le promoteur d'apoptose abt-263
WO2009156737A1 (fr) 2008-06-25 2009-12-30 Vernalis (R&D) Limited Dérivés de triazalo [4, 5-d] pyramidine et leur utilisation comme antagonistes des récepteurs de la purine
US8329754B2 (en) 2008-08-04 2012-12-11 Novartis Ag Squaramide derivatives as CXCR2 antagonist
US7989497B2 (en) 2008-08-04 2011-08-02 Novartis Ag Squaramide derivatives as CXCR2 antagonist
US20110251205A1 (en) 2008-08-04 2011-10-13 Novartis Ag Squaramide derivatives as cxcr2 antagonist
US8288588B2 (en) 2008-08-04 2012-10-16 Novartis Ag Squaramide derivatives as CXCR2 antagonist
US20110251206A1 (en) 2008-08-04 2011-10-13 Novartis Ag Squaramide derivatives as CXCR2 antagonist
US8722925B2 (en) 2008-08-04 2014-05-13 Novartis Ag Squaramide derivatives as CXCR2 antagonist
WO2010015613A1 (fr) 2008-08-04 2010-02-11 Novartis Ag Composés organiques
US9115087B2 (en) 2008-08-04 2015-08-25 Novartis Ag Squaramide derivatives as CXCR2 antagonist
WO2010019570A2 (fr) 2008-08-11 2010-02-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
US8415355B2 (en) 2008-08-22 2013-04-09 Novartis Ag Pyrrolopyrimidine compounds and their uses
US8685980B2 (en) 2008-08-22 2014-04-01 Novartis Ag Pyrrolopyrimidine compounds and their uses
WO2010020675A1 (fr) 2008-08-22 2010-02-25 Novartis Ag Composés de pyrrolopyrimidine et leurs utilisations
US8609089B2 (en) 2008-08-25 2013-12-17 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US20120114649A1 (en) 2008-08-25 2012-05-10 Amplimmune, Inc. Delaware Compositions of pd-1 antagonists and methods of use
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
US8114845B2 (en) 2008-08-25 2012-02-14 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US8927697B2 (en) 2008-09-12 2015-01-06 Isis Innovation Limited PD-1 specific antibodies and uses thereof
WO2010033866A2 (fr) 2008-09-19 2010-03-25 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Anticorps monoclonaux de cspg4 utilises dans le diagnostic et le traitement du carcinome mammaire de type basal
US20140004124A1 (en) 2008-09-19 2014-01-02 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Monoclonal antibodies for cspg4 for the diagnosis and treatment of basal breast carcinoma
US9102727B2 (en) 2008-09-26 2015-08-11 Emory University Human anti-PD-1 antibodies and uses therefor
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
WO2010063802A1 (fr) 2008-12-05 2010-06-10 Novartis Ag Cyclobutène-1,2-diones 3,4-disubstituées en tant qu'antagonistes de récepteur cxcr2
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US9029393B2 (en) 2009-01-26 2015-05-12 Kaldi Pharma, Sas Adenosine receptor ligands and uses thereof
US20120039906A1 (en) 2009-02-09 2012-02-16 INSER (Institut National de la Recherche Medicale) PD-1 Antibodies and PD-L1 Antibodies and Uses Thereof
US9133197B2 (en) 2009-03-20 2015-09-15 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Oxidated derivatives of triazolylpurines useful as ligands of the adenosine A2A receptor and their use as medicaments
WO2010120561A1 (fr) 2009-04-01 2010-10-21 Genentech, Inc. Anticorps et immunoconjugués anti-fcrh5 et procédés d'utilisation
WO2010114940A1 (fr) 2009-04-01 2010-10-07 Genentech, Inc. Anticorps anti-fcrh5 et immunoconjugués et procédés d'utilisation
WO2010129304A2 (fr) 2009-04-27 2010-11-11 Oncomed Pharmaceuticals, Inc. Procédé de fabrication de molécules hétéromultimères
WO2010126066A1 (fr) 2009-04-27 2010-11-04 協和発酵キリン株式会社 Anticorps anti-il-3rα destiné à être utilisé dans le traitement d'hématomes
WO2011049677A1 (fr) 2009-09-02 2011-04-28 Irm Llc Composés et compositions utilisés en tant que modulateurs de l'activité tlr
US8709424B2 (en) 2009-09-03 2014-04-29 Merck Sharp & Dohme Corp. Anti-GITR antibodies
WO2011028683A1 (fr) 2009-09-03 2011-03-10 Schering Corporation Anticorps anti-gitr
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US8779108B2 (en) 2009-11-24 2014-07-15 Medimmune, Limited Targeted binding agents against B7-H1
US20110268656A1 (en) 2009-12-02 2011-11-03 David Ho J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
WO2011076786A1 (fr) 2009-12-22 2011-06-30 Novartis Ag Isoquinolinones et quinazolinones substituées
WO2011076922A1 (fr) 2009-12-23 2011-06-30 Synimmune Gmbh Anticorps anti-flt3 et leurs méthodes d'emploi
WO2011095625A1 (fr) 2010-02-05 2011-08-11 Heptares Therapeutics Limited Dérivés de 1,2,4-triazine-4-amine
US20120009181A1 (en) 2010-02-24 2012-01-12 Ab Olga Folate Receptor 1 Antibodies and Immunoconjugates and Uses Thereof
US8993731B2 (en) 2010-03-11 2015-03-31 Ucb Biopharma Sprl PD-1 antibody
US8796284B2 (en) 2010-03-31 2014-08-05 Palobiofarma, S.L. 4-aminopyrimidine derivatives and their as as adenosine A2a receptor antagonists
WO2011131746A2 (fr) 2010-04-20 2011-10-27 Genmab A/S Protéines contenant des anticorps fc hétérodimères et leurs procédés de production
WO2011156518A2 (fr) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Modulateur du récepteur oestrogénique et utilisation de ces derniers
US8552156B2 (en) 2010-06-11 2013-10-08 Kyowa Hakko Kirin Co., Ltd Anti-TIM-3 antibody
US20130101607A1 (en) 2010-06-15 2013-04-25 Thomas J. Kipps Receptor Tyrosine Kinase-Like Orphan Receptor 1 (ROR1) Single Chain FV Antibody Fragment Conjugates and Methods of Use Thereof
WO2011159847A2 (fr) 2010-06-15 2011-12-22 The Regents Of The University Of California Conjugués du fragment d'anticorps fv à chaîne unique dirigé contre le récepteur orphelin 1 analogue au récepteur à la tyrosine kinase (ror1) et leurs procédés d'utilisation
WO2011159769A2 (fr) 2010-06-17 2011-12-22 Aragon Pharmaceuticals, Inc. Modulateurs de récepteur d'œstrogène d'indane et utilisations de ceux-ci
US9163087B2 (en) 2010-06-18 2015-10-20 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against TIM-3 and PD-1 for immunotherapy in chronic immune conditions
WO2011160119A2 (fr) 2010-06-19 2011-12-22 Memorial Sloan-Kettering Cancer Center Anticorps contre gd2
US8907053B2 (en) 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
WO2012033885A1 (fr) 2010-09-08 2012-03-15 Baylor College Of Medicine Immunothérapie des cancers utilisant des lymphocytes t génétiquement modifiés, spécifiques de gd2
WO2012037411A2 (fr) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Modulateurs des récepteurs des oestrogènes et leurs utilisations
WO2012037410A2 (fr) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Modulateurs des récepteurs des oestrogènes et leurs utilisations
US20120071535A1 (en) 2010-09-16 2012-03-22 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2012062713A1 (fr) 2010-11-08 2012-05-18 Novartis Ag Polypeptides se liant aux récepteurs de chimiokines
WO2012079000A1 (fr) 2010-12-09 2012-06-14 The Trustees Of The University Of Pennsylvania Utilisation de lymphocytes t modifiés par un récepteur chimérique d'antigènes chimérique pour traiter le cancer
US8362211B2 (en) 2010-12-30 2013-01-29 Takeda Pharmaceutical Company Limited Anti-CD38 antibodies
WO2012135854A2 (fr) 2011-04-01 2012-10-04 Memorial Sloan-Kettering Cancer Center Anticorps contre des peptides cytosoliques
WO2012145493A1 (fr) 2011-04-20 2012-10-26 Amplimmune, Inc. Anticorps et autres molécules qui se lient à b7-h1 et à pd-1
US9205148B2 (en) 2011-04-20 2015-12-08 Medimmune, Llc Antibodies and other molecules that bind B7-H1 and PD-1
US20120288506A1 (en) 2011-05-12 2012-11-15 Imclone Llc C-kit antibodies and uses thereof
WO2012163805A1 (fr) 2011-05-27 2012-12-06 Glaxo Group Limited Protéines de liaison à bcma (cd269/tnfrsf17)
WO2012167143A1 (fr) 2011-06-03 2012-12-06 Xoma Technology Ltd. Anticorps spécifiques du tgf bêta
WO2012175222A1 (fr) 2011-06-24 2012-12-27 Cytune Immunocytokines à base d'il-15 et domaine sushi d'il-15rα
US8841418B2 (en) 2011-07-01 2014-09-23 Cellerant Therapeutics, Inc. Antibodies that specifically bind to TIM3
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013030803A1 (fr) 2011-09-02 2013-03-07 Novartis Ag Sel de choline d'un composé anti-inflammatoire à base de cyclobutènedione substitué
WO2013040557A2 (fr) 2011-09-16 2013-03-21 The Trustees Of The University Of Pennsylvania Lymphocytes t à arn modifié pour le traitement du cancer
WO2013040371A2 (fr) 2011-09-16 2013-03-21 Baylor College Of Medicine Ciblage du microenvironnement tumoral au moyen de cellules nkt modifiées
WO2013061273A1 (fr) 2011-10-25 2013-05-02 Massimo Dominici Cellule effectrice modifiée (ou récepteur chimérique) pour traiter la néoplasie exprimant le disialoganglioside gd2
WO2013060867A2 (fr) 2011-10-27 2013-05-02 Genmab A/S Production de protéines hétérodimères
WO2013063419A2 (fr) 2011-10-28 2013-05-02 The Trustees Of The University Of Pennsylvania Récepteur immunitaire chimérique spécifique complètement humain, anti-mésothéline pour un ciblage redirigé de cellules exprimant la mésothéline
WO2013074916A1 (fr) 2011-11-18 2013-05-23 Board Of Regents, The University Of Texas System Lymphocytes t car+ génétiquement modifiés pour éliminer l'expression du récepteur des lymphocytes t et/ou le système hla
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
WO2013085552A1 (fr) 2011-12-08 2013-06-13 Cleveland Clinic Foundation Fixation de la voûte glénoïde
WO2013111105A1 (fr) 2012-01-26 2013-08-01 Novartis Ag Composés imidazopyrrolidinone
WO2013123061A1 (fr) 2012-02-13 2013-08-22 Seattle Children's Hospital D/B/A Seattle Children's Research Institute Récepteurs d'antigène chimères bispécifiques et utilisations thérapeutiques de ceux-ci
WO2013126712A1 (fr) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Topicompositions et procédés pour produire une population de lymphocytes t tenaces utiles dans le traitement du cancer
WO2013142034A1 (fr) 2012-03-23 2013-09-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Récepteurs d'antigène chimérique anti-mésothéline
US20130295007A1 (en) 2012-05-01 2013-11-07 Genentech, Inc. Anti-pmel17 antibodies and immunoconjugates
WO2013165940A1 (fr) 2012-05-01 2013-11-07 Genentech, Inc. Anticorps anti-pmel17 et immunoconjugués
WO2013168108A2 (fr) 2012-05-09 2013-11-14 Novartis Ag Polypeptides de liaison de récepteur de chimiokine
WO2013173820A2 (fr) 2012-05-18 2013-11-21 Scott & White Healthcare Immunofusion bispécifique (ifb) de scfv
WO2013179174A1 (fr) 2012-05-29 2013-12-05 Koninklijke Philips N.V. Système d'éclairage
US9175082B2 (en) 2012-05-31 2015-11-03 Sorrento Therapeutics, Inc. Antigen binding proteins that bind PD-L1
WO2013192294A1 (fr) 2012-06-20 2013-12-27 Boston 3T Biotechnologies, Inc. Thérapies cellulaires pour le traitement et la prévention de cancers et d'autres troubles du système immunitaire
US9505839B2 (en) 2012-07-02 2016-11-29 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (LAG-3), and uses thereof
WO2014022758A1 (fr) 2012-08-03 2014-02-06 Dana-Farber Cancer Institute, Inc. Anticorps de liaison double à agent unique anti-pd-l1 et pd-l2 et procédés d'utilisation
WO2014031687A1 (fr) 2012-08-20 2014-02-27 Jensen, Michael Procédé et compositions pour l'immunothérapie cellulaire
US20140086932A1 (en) 2012-09-17 2014-03-27 Peter G. Traber Method for enhancing specific immunotherapies in cancer treatment
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
WO2014066527A2 (fr) 2012-10-24 2014-05-01 Admune Therapeutics Llc Formes d'il-15r alpha, cellules exprimant des formes d'il-15r alpha, et utilisations thérapeutiques d'il-15r alpha et de complexes il-15/il-15r alpha
WO2014089335A2 (fr) 2012-12-07 2014-06-12 Amgen Inc. Protéines de liaison à l'antigène bcma
WO2014100079A1 (fr) 2012-12-21 2014-06-26 Merck Sharp & Dohme Corp. Anticorps qui se lient au ligand 1 de la mort programmée humaine (pd-l1)
WO2014122144A1 (fr) 2013-02-05 2014-08-14 Engmab Ag Anticorps bispécifiques anti-cd3ɛ et bcma
WO2014130310A1 (fr) 2013-02-19 2014-08-28 Novartis Ag Dérivés de benzothiophène et compositions correspondantes en tant qu'agents de dégradation sélectifs des récepteurs des œstrogènes
WO2014130635A1 (fr) 2013-02-20 2014-08-28 Novartis Ag Ciblage efficace de la leucémie primaire humaine au moyen de lymphocytes t génétiquement modifiés des récepteurs d'antigènes chimériques anti-cd123
WO2014130657A1 (fr) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Traitement du cancer au moyen d'un récepteur d'antigènes chimériques anti-egfrviii humanisés
WO2014138805A1 (fr) 2013-03-14 2014-09-18 Csl Limited Agents anti-il-3r alpha et leurs utilisations
WO2014138819A1 (fr) 2013-03-14 2014-09-18 Csl Limited Agents qui neutralisent la signalisation par il-3 et leurs utilisations
WO2014140180A1 (fr) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Protéines de liaison anti-lag-3
WO2014144622A2 (fr) 2013-03-15 2014-09-18 Stephen Forman Lymphocytes t redirigés par des récepteurs d'antigènes chimériques spécifiques de cd123 et leurs procédés d'utilisation
WO2014140248A1 (fr) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Molécules liantes pour bcma et cd3
WO2014153270A1 (fr) 2013-03-16 2014-09-25 Novartis Ag Traitement du cancer à l'aide d'un récepteur d'antigène chimérique anti-cd19 humanisé
WO2014165707A2 (fr) 2013-04-03 2014-10-09 Memorial Sloan-Kettering Cancer Center Génération efficace de lymphocytes t ciblant une tumeur dérivés de cellules souches pluripotentes
WO2014179664A2 (fr) 2013-05-02 2014-11-06 Anaptysbio, Inc. Anticorps dirigés contre la protéine de mort programmée 1 (pd-1)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2014210064A1 (fr) 2013-06-24 2014-12-31 Genentech, Inc. Anticorps anti-fcrh5
WO2014209804A1 (fr) 2013-06-24 2014-12-31 Biomed Valley Discoveries, Inc. Anticorps bispécifiques
WO2015026684A1 (fr) 2013-08-20 2015-02-26 Merck Sharp & Dohme Corp. Modulation d'immunité tumorale
WO2015031667A2 (fr) 2013-08-30 2015-03-05 Amgen Inc. Protéines de liaison à l'antigène gitr
US9464139B2 (en) 2013-08-30 2016-10-11 Amgen Inc. GITR antigen binding proteins and methods of use thereof
US8735553B1 (en) 2013-09-13 2014-05-27 Beigene, Ltd. Anti-PD1 antibodies and their use as therapeutics and diagnostics
WO2015061668A1 (fr) 2013-10-25 2015-04-30 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux anti-pd-l1 et fragments de ceux-ci
WO2015081158A1 (fr) 2013-11-26 2015-06-04 Bristol-Myers Squibb Company Procédé de traitement du vih par perturbation de la signalisation pd-1/pd-l1
WO2015085847A1 (fr) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Anticorps anti-pd-1, son fragment de liaison à l'antigène, et son application médicale
WO2015090230A1 (fr) 2013-12-19 2015-06-25 Novartis Ag Récepteurs antigéniques chimériques de la mésothéline humaine et leurs utilisations
WO2015109124A2 (fr) 2014-01-15 2015-07-23 Kadmon Corporation, Llc Agents immunomodulateurs
US20170015758A1 (en) 2014-01-21 2017-01-19 Medimmune, Llc Compositions And Methods For Modulating And Redirecting Immune Responses
WO2015112800A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains se liant à pd-1
WO2015112805A1 (fr) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Anticorps humains dirigés contre pd-l1
US20150210769A1 (en) 2014-01-24 2015-07-30 Novartis Ag Antibody molecules to pd-1 and uses thereof
WO2015116539A1 (fr) 2014-01-28 2015-08-06 Bristol-Myers Squibb Company Anticorps anti-lag-3 pour traiter des hémopathies malignes
US20150218274A1 (en) 2014-01-31 2015-08-06 Novartis Ag Antibody molecules to tim-3 and uses thereof
WO2015136017A1 (fr) 2014-03-13 2015-09-17 F. Hoffmann-La Roche Ag Méthodes et compositions pour moduler des mutants du récepteur des oestrogènes
US20150259420A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
WO2015142675A2 (fr) 2014-03-15 2015-09-24 Novartis Ag Traitement du cancer au moyen d'un récepteur antigénique chimérique
WO2015184099A1 (fr) 2014-05-28 2015-12-03 4-Antibody Ag Anticorps anti-gitr et leurs procédés d'utilisation
US20150368349A1 (en) 2014-05-28 2015-12-24 4-Antibody Ag Anti-GITR Antibodies and Methods of Use Thereof
WO2015181342A1 (fr) 2014-05-29 2015-12-03 Spring Bioscience Corporation Anticorps dirigés contre pd-l1 et leurs utilisations
US9228016B2 (en) 2014-06-06 2016-01-05 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
WO2015195163A1 (fr) 2014-06-20 2015-12-23 R-Pharm Overseas, Inc. Anticorps totalement humain anti-pd-l1
WO2015200119A1 (fr) 2014-06-26 2015-12-30 Macrogenics, Inc. Dianticorps liés par covalence, présentant une immunoréactivité avec pd-1 et lag-3 et leurs procédés d'utilisation
WO2016000619A1 (fr) 2014-07-03 2016-01-07 Beigene, Ltd. Anticorps anti-pd-l1 et leur utilisation comme agents thérapeutiques et diagnostiques
WO2016014535A1 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer au moyen d'un récepteur d'antigènes chimériques cll-1
WO2016014565A2 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer au moyen d'un récepteur d'antigène chimérique anti-bcma humanisé
WO2016014576A1 (fr) 2014-07-21 2016-01-28 Novartis Ag Traitement du cancer à l'aide du récepteur antigénique chimérique anti-cd33
WO2016014789A2 (fr) 2014-07-24 2016-01-28 Bluebird Bio, Inc. Récepteurs de l'antigène chimérique bcma
WO2016028896A1 (fr) 2014-08-19 2016-02-25 Novartis Ag Récepteur d'antigène chimérique anti-cd123 (car) utilisé dans le traitement du cancer
WO2016028672A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps et fragments de fixation à l'antigène anti-lag3
WO2016054638A1 (fr) 2014-10-03 2016-04-07 Dana-Farber Cancer Institute, Inc. Anticorps dirigés contre le récepteur du facteur de nécrose tumorale induit par glucocorticoïdes (gitr) et leurs procédés d'utilisation
WO2016057846A1 (fr) 2014-10-08 2016-04-14 Novartis Ag Compositions et procédés d'utilisation pour une réponse immunitaire accrue et traitement contre le cancer
US20160108123A1 (en) 2014-10-14 2016-04-21 Novartis Ag Antibody molecules to pd-l1 and uses thereof
WO2016071448A1 (fr) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Anticorps anti-tim3 et procédés d'utilisation
US20160129108A1 (en) 2014-11-11 2016-05-12 Medimmune Limited Therapeutic combinations comprising anti-cd73 antibodies and uses thereof
WO2016092419A1 (fr) 2014-12-09 2016-06-16 Rinat Neuroscience Corp. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
WO2016111947A2 (fr) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Anticorps inhibiteurs d'interactions de tim-3:lilrb2 et leurs utilisations
WO2016144803A2 (fr) 2015-03-06 2016-09-15 Sorrento Therapeutics, Inc. Anticorps thérapeutiques se liant à tim3
WO2016161270A1 (fr) 2015-04-01 2016-10-06 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de cellule t et protéine 3 de mucine (tim-3)
WO2016196792A1 (fr) 2015-06-03 2016-12-08 Bristol-Myers Squibb Company Anticorps anti-gitr pour le diagnostic du cancer
US20170022284A1 (en) 2015-07-23 2017-01-26 Inhibrx Lp Multivalent and multispecific gitr-binding fusion proteins
WO2017015623A2 (fr) 2015-07-23 2017-01-26 Inhibrx Lp Protéines hybrides multivalentes et multispécifiques se liant à gitr
WO2017025918A1 (fr) 2015-08-11 2017-02-16 Novartis Ag 5-bromo -2,6-di- (1h-pyrazol-1-yl)pyrimidin-4-amine pour utilisation dans le traitement du cancer
WO2017025610A1 (fr) 2015-08-12 2017-02-16 Medimmune Limited Protéines de fusion gitrl et leurs utilisations
US20170073386A1 (en) 2015-08-12 2017-03-16 Medimmune Limited Gitrl fusion proteins and uses thereof
WO2017027645A1 (fr) 2015-08-13 2017-02-16 Merck Sharp & Dohme Corp. Composés di-nucléotidiques cycliques en tant qu'agonistes de sting
US20180099940A1 (en) * 2016-10-11 2018-04-12 Arvinas, Inc. Compounds and methods for the targeted degradation of androgen receptor
WO2018102725A1 (fr) * 2016-12-01 2018-06-07 Arvinas, Inc. Dérivés de tétrahydronaphtalène et de tétrahydroisoquinoléine en tant qu'agents de dégradation des récepteurs des œstrogènes
WO2018140809A1 (fr) * 2017-01-26 2018-08-02 Arvinas, Inc. Modulateurs du récepteur des œstrogènes de protéolyse et procédés d'utilisation associés
WO2019038717A1 (fr) 2017-08-23 2019-02-28 Novartis Ag Dérivés de 3-(1-oxoisoindolin-2-yl)pipéridine-2,6-dione et leurs utilisations
WO2019079569A1 (fr) * 2017-10-18 2019-04-25 Novartis Ag Compositions et méthodes pour la dégradation sélective d'une protéine

Non-Patent Citations (134)

* Cited by examiner, † Cited by third party
Title
"4507 Pre-Clinical Characterization of T Cell-Dependent Bispecific Antibody Anti-CD79b/CD3 As a Potential Therapy for B Cell Malignancies", 56TH ASH ANNUAL MEETING AND EXPOSITION, 6 December 2014 (2014-12-06)
"Design of Prodrugs", 1985, ELSEVIER, article "Design and Applications of Prodrugs"
"Pro-Drugs as Novel Delivery Systems", 1975, AM. CHEM. SOC.
16TH ANNU MEET AM SOC GEN CELL THER (ASGCT, 15 May 2013 (2013-05-15)
ABENGOZAR ET AL., BLOOD, vol. 119, no. 19, 2012, pages 4565 - 4576
ACHYUT ET AL., GASTROENTEROLOGY, vol. 141, no. 4, 2011, pages 1167 - 78
AGGEN ET AL., GENE THER., vol. 19, no. 4, 2012, pages 365 - 74
AIGNER ET AL., LEUKEMIA, vol. 27, no. 5, 2013, pages 1107 - 1115
APTE ET AL., ADV. EXP. MED. BIOL., 2000, pages 277 - 88
APTE ET AL., CANCER METASTASIS REV., 2006, pages 387 - 408
ASANUMA, S. ET AL., CANCER SCI., vol. 104, 2013, pages 1097 - 1106
BEJCEK, CANCER RESEARCH, vol. 55, 1995, pages 2346 - 178,949-982
BIN DHUBAN, K. ET AL., J. IMMUNOL., vol. 194, 2015, pages 3687 - 96
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
BLOBE ET AL., N ENGL J MED., vol. 342, no. 18, 2000, pages 1350 - 8
BOER K. ET AL., THERAPEUTIC ADVANCES IN MEDICAL ONCOLOGY, vol. 9, no. 7, 2017, pages 465 - 479
BRASS ET AL., CLIN CANCER RES, vol. 7, no. 6, 2001, pages 1490 - 1496
BRENTJENS ET AL., BLOOD, vol. 118, no. 18, 2011, pages 4817 - 4828
BROOKS ET AL., PNAS, vol. 107, no. 22, 2010, pages 10056 - 10061
CARON ET AL., CANCER RES, vol. 52, no. 24, 1992, pages 6761 - 6767
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 1204531-25-80
CHEUNG ET AL., CANCER RES, vol. 45, no. 6, 1985, pages 2642 - 2649
CHEUNG ET AL., J CLIN ONCOL, vol. 16, no. 9, 1998, pages 3053 - 3060
CHEUNG ET AL., J CLIN ONCOL, vol. 5, no. 9, 1987, pages 1430 - 1440
CHINNASAMY ET AL., J CLIN INVEST, vol. 120, no. 11, 2010, pages 3953 - 3968
CHMIELEWSKI ET AL., GASTOENTEROLOGY, vol. 143, no. 4, 2012, pages 1095 - 1107
DAO ET AL., SCI TRANSL MED, vol. 5, no. 176, 2013, pages 176ra33
DE GROOT ET AL., J IMMUNOL, vol. 183, no. 6, 2009, pages 4127 - 4134
DEMBIC ET AL., NATURE, vol. 320, 1986, pages 232 - 238
DINARELLO, EUR. J. IMMUNOL., 2010, pages 599 - 606
DOLEZAL ET AL., PROTEIN ENGINEERING, vol. 16, no. 1, 2003, pages 47 - 56
DORNAN ET AL.: "Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma", BLOOD., vol. 114, no. 13, 24 September 2009 (2009-09-24), pages 2721 - 9, XP055075268, DOI: 10.1182/blood-2009-02-205500
DUTOUR ET AL., ADV HEMATOL, vol. 2012, 2012, pages 683065
E.L. ELIELS.H. WILENL.N. MANDER: "Stereochemistry of Organic Compounds", 1994, WILEY-INTERSCIENCE
ELKINS ET AL.: "FcRL5 as a target of antibody-drug conjugates for the treatment of multiple myeloma", MOL CANCER THER., vol. 1 1, no. 10, October 2012 (2012-10-01), pages 2222 - 32, XP055141107, DOI: 10.1158/1535-7163.MCT-12-0087
ENGELS ET AL., HUM. GENE THER., vol. 16, 2005, pages 799 - 810
FENG ET AL.: "Glypican-3 antibodies: a new therapeutic target for liver cancer", FEBS LETT., vol. 588, no. 2, 21 January 2014 (2014-01-21), pages 377 - 82, XP028669969, DOI: 10.1016/j.febslet.2013.10.002
FINN ET AL., BREAST CANCER RESEARCH, vol. 11, no. 5, 2009, pages R77
FRIGERIO ET AL., EUROPEAN J CANCER, vol. 49, no. 9, 2013, pages 2223 - 2232
GARNER F ET AL., ANTICANCER DRUGS, vol. 26, no. 9, 2015, pages 948 - 56
GETNET, D. ET AL., MOL. IMMUNOL., vol. 47, 2010, pages 1595 - 1600
GHODS ET AL., BIOTECHNOL APPL BIOCHEM, 2013
GORDONBREACH: "A Textbook of Drug Design and Development", 1991
HAMID, O. ET AL., NEW ENGLAND JOURNAL OF MEDICINE, vol. 369, no. 2, 2013, pages 134 - 44
HANDGRETINGER ET AL., CANCER IMMUNOL IMMUNOTHER, vol. 35, no. 3, 1992, pages 199 - 204
HOFHEINZ ET AL., ONCOLOGY RESEARCH AND TREATMENT, vol. 26, no. 1, 2003
HOLLINGER ET AL., PROC NATL ACAD. SCI. U.S.A., vol. 90, 1993, pages 6444 - 6448
HOMBACH ET AL., GASTROENTEROLOGY, vol. 113, no. 4, 1997, pages 1163 - 1170
HONG ET AL., J IMMUNOTHER, vol. 37, no. 2, 2014, pages 93 - 104
HUANG ET AL., CANCER RES, vol. 72, no. 1, 2012, pages 271 - 281
HUDECEK ET AL., CLIN CANCER RES, vol. 19, no. 12, 2013, pages 3153 - 3164
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
JAGER ET AL., APPL IMMUNOHISTOCHEM MOL MORPHOL, vol. 15, no. 1, 2007, pages 77 - 83
JOHN, L.B. ET AL., MOL. IMMUNOL., vol. 48, 2011, pages 1272 - 1278
JUNE ET AL., NATURE REVIEWS IMMUNOLOGY, vol. 9.10, 2009, pages 704 - 716
KATAOKA, D. ET AL., NATURE GENETICS, vol. 47, 2015, pages 1304 - 1315
KATO ET AL., LEUK RES, vol. 37, no. 1, 2013, pages 83 - 88
KELLY ET AL., CANCER BIOTHER RADIOPHARM, vol. 23, no. 4, 2008, pages 411 - 423
KERSHAW ET AL., NAT. REV. IMMUNOL., vol. 5, 2005, pages 928 - 940
KIM, H-J. ET AL., SCIENCE, vol. 350, 2015, pages 334 - 339
KMIECIK ET AL., ONCOIMMUNOLOGY, vol. 3, no. l, 2014, pages e27185
KOCHENDERFER ET AL., BLOOD, vol. 122, no. 25, 2013, pages 4129 - 2973
KOCHENDERFER, J.N. ET AL., BLOOD, vol. 116, no. 20, 2010, pages 4099 - 1262
KOCHENDERFER, J.N. ET AL., J. IMMUNOTHER., vol. 32, no. 7, 2009, pages 689 - 702
KUDRYASHOV V ET AL., GLYCOCONJ J., vol. 15, no. 3, 1998, pages 243 - 9
LANZAVECCHIA ET AL., EUR. J. IMMUNOL., vol. 17, 1987, pages 105
LAPUSAN ET AL., INVEST NEW DRUGS, vol. 30, no. 3, 2012, pages 1121 - 1131
LEWITT, ANNALS OF NEUROLOGY, vol. 63, no. 3, 2008, pages 295 - 302
LOU ET AL., PROC NATL ACAD SCI USA, vol. 111, no. 7, 2014, pages 2482 - 2487
LUO ET AL., EMBO MOL. MED., vol. 4, no. 6, 2012, pages 453 - 461
MAECKER ET AL., BLOOD, vol. 102, no. 9, 2003, pages 3287 - 3294
MAHNE ET AL., CANCER RES., vol. 77, no. 5, 2017, pages 1108 - 1118
MALIAR ET AL., GASTROENTEROLOGY, vol. 143, no. 5, 2012, pages 1375 - 1384
MARTY ET AL., CANCER LETT, vol. 235, no. 2, 2006, pages 298 - 308
MBRL: BREMER E-G ET AL., J BIOL CHEM, vol. 259, 1984, pages 14773 - 14777
MCDONELL ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, no. 12, 2015, pages 4883 - 4887
MILLER ET AL., BMC PHARMACOLOGY AND TOXICOLOGY, vol. 16, 2015, pages 18
MILLER ET AL., EUR J DRUG METAB PHARMACOKINET, vol. 39, 2014, pages 173 - 181
MILLER ET AL., J. IMMUNOL., 2007, pages 6933 - 42
MINO-KENUDSON ET AL., CLIN CANCER RES, vol. 16, no. 5, 2010, pages 1561 - 1571
MORGAN ET AL., J. IMMUNOL., vol. 171, 2003, pages 3287 - 3295
MORGENROTH ET AL., PROSTATE, vol. 67, no. 10, 2007, pages 1121 - 1131
MUJOO ET AL., CANCER RES., vol. 47, no. 4, 1987, pages 1098 - 1104
MURPHY ET AL., IMMUNITY, vol. 22, 2005, pages 403 - 414
MYERSUCKUN: "An anti-CD72 immunotoxin against therapy-refractory B-lineage acute lymphoblastic leukemia", LEUK LYMPHOMA., vol. 18, no. 1-2, June 1995 (1995-06-01), pages 119 - 22, XP002964114
NAGAE ET AL., J BIOL CHEM, vol. 288, no. 47, 2013, pages 33784 - 33796
NAKANO KISHIGURO TKONISHI H ET AL.: "Generation of a humanized anti-glypican 3 antibody by CDR grafting and stability optimization", ANTICANCER DRUGS, vol. 21, no. 10, November 2010 (2010-11-01), pages 907 - 916, XP008177208, DOI: 10.1097/CAD.0b013e32833f5d68
NAKAWAGA, H. ET AL., PNAS, vol. 113, 2016, pages 6248 - 6253
NEESON ET AL., J IMMUNOL, vol. 190, May 2013 (2013-05-01), pages 177.10
NEJATOLLAHI ET AL., J OF ONCOLOGY, vol. 2013, 2013
NING ET AL., MOL CANCER THER., vol. 11, no. 6, 2012, pages 1353 - 64
NOORDHUIS ET AL.: "Targeting of CLEC12A In Acute Myeloid Leukemia by Antibody-Drug-Conjugates and Bispecific CLL-lxCD3 BiTE Antibody", 53RD ASH ANNUAL MEETING AND EXPOSITION, 10 December 2011 (2011-12-10)
OSTERMANN ET AL., CLINICAL CANCER RESEARCH, vol. 14, 2008, pages 4584 - 4592
PARKER ET AL., PROTEIN EXPR PURIF, vol. 89, no. 2, 2013, pages 136 - 145
PERDOMO, J. ET AL., J. BIOL. CHEM., vol. 275, 2000, pages 38347 - 38354
PETER H SCHAFER ET AL: "Cereblon modulator iberdomide induces degradation of the transcription factors Ikaros and Aiolos: immunomodulation in healthy volunteers and relevance to systemic lupus erythematosus", ANNALS OF THE RHEUMATIC DISEASES, vol. 77, no. 10, 26 June 2018 (2018-06-26), GB, pages 1516 - 1523, XP055686777, ISSN: 0003-4967, DOI: 10.1136/annrheumdis-2017-212916 *
PIZZITOLA ET AL., LEUKEMIA, 2014
POISON ET AL.: "Antibody-Drug Conjugates for the Treatment of Non-Hodgkin's Lymphoma: Target and Linker-Drug Selection", CANCER RES, vol. 69, 15 March 2009 (2009-03-15), pages 2358, XP055076856, DOI: 10.1158/0008-5472.CAN-08-2250
PONTE J, CLINICAL IMMUNOLOGY, vol. 135, 2010, pages S96
ROSS ET AL., CANCER RES, vol. 76, no. 14, 2016
ROSSIG ET AL., MOL. THER., vol. 10, 2004, pages 5 - 18
ROSZKOWSKI ET AL., CANCER RES., vol. 65, 2005, pages 1570 - 1576
S.M. BIRGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
SALOMONSSON E ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 286, no. 16, 2011, pages 13801 - 13804
SCHUMACHER, NAT. REV. IMMUNOL.,, vol. 2, 2002, pages 512 - 519
SCOTT AM ET AL., CANCER RES, vol. 60, 2000, pages 3254 - 61
SMITH ET AL.: "Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement", CLINICAL & TRANSLATIONAL IMMUNOLOGY, vol. 4, 2015, pages e31
SONG ET AL., MED ONCOL, vol. 29, no. 4, 2012, pages 2923 - 2931
SONG ET AL., TARGET ONCOL, 14 August 2013 (2013-08-14)
STONE ET AL., ONCOLMMUNOLOGY, vol. 1, no. 6, 2012, pages 863 - 873
T.E. BEESLEYR.P.W. SCOTT: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
TAI ET AL., BLOOD, vol. 112, no. 4, 2008, pages 1329 - 37
THEMELI M. ET AL., NAT. BIOTECHNOL., vol. 31, 2013, pages 928 - 933
TIAN ET AL., CELL SIGNAL., vol. 23, no. 6, 2011, pages 951 - 62
TOBIAS MAETZIG ET AL.: "Gammaretroviral Vectors: Biology, Technology and Application", VIRUSES, vol. 3, no. 6, June 2011 (2011-06-01), pages 677 - 713
TORRES-GUZMAN R ET AL., ONCOTARGET, 2017
TRAN ET AL., J EXP MED, vol. 210, no. 6, 2013, pages 1125 - 1135
TRUTYURRUTIA, PANEREATOLOGY., vol. 7, no. 5-6, 2007, pages 423 - 35
VORONOV ET AL., FRONT PHYSIOL., 2014, pages 114
VORONOV ET AL., PROC. NATL. ACAD. SCI. U.S.A., 2003, pages 2645 - 50
WAKEFIELDHILL, NAT REV CANCER., vol. 13, no. 5, 2013, pages 328 - 41
WANG ET AL., J EXP MED., vol. 184, no. 6, 1996, pages 2207 - 16
WILLEMSEN ET AL., J IMMUNOL, vol. 174, no. 12, 2005, pages 7853 - 7858
WILLEMSEN RA ET AL., GENE THERAPY, vol. 7, 2000, pages 1369 - 1377
WOJTOWICZ-PRAGA, LNVEST NEW DRUGS., vol. 21, no. 1, 2003, pages 21 - 32
XU ET AL., LEUK LYMPHOMA, vol. 54, no. 2, 2012, pages 255 - 260
XUE ET AL., CLIN. EXP. IMMUNOL., vol. 139, 2005, pages 167 - 172
YANG ET AL., TRENDS IMMUNOL., vol. 31, no. 6, 2010, pages 220 - 7
YU ET AL., MOL THER, vol. 22, no. 1, 2014, pages 102 - 111
ZARBOCK ET AL., BR J PHARMACOL., vol. 155, no. 3, 2008, pages 357 - 64
ZHANG T ET AL., CANCER GENE THER, vol. 11, 2004, pages 487 - 496
ZHANG, Z. ET AL., BLOOD, vol. 110, no. 5, 2007, pages 1656 - 2197
ZHAO ET AL., J IMMUNOL METHODS, vol. 363, no. 2, 2011, pages 221 - 232
ZHAO ET AL., J. IMMUNOL., vol. 174, 2005, pages 4415 - 4423

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112574094A (zh) * 2020-12-14 2021-03-30 成都大学 吲哚酮衍生物及其制药用途
CN112574094B (zh) * 2020-12-14 2022-07-01 成都大学 吲哚酮衍生物及其制药用途
WO2022216573A1 (fr) * 2021-04-05 2022-10-13 Bristol-Myers Squibb Company Composés d'oxoisoindoline à substitution pyridinyle pour le traitement du cancer
WO2022216644A1 (fr) * 2021-04-06 2022-10-13 Bristol-Myers Squibb Company Composés oxoisoindoline substitués par pyridinyle
US11718601B2 (en) 2021-04-06 2023-08-08 Bristol-Myers Squibb Company Pyridinyl substituted oxoisoindoline compounds
US11878968B2 (en) 2021-07-09 2024-01-23 Plexium, Inc. Aryl compounds and pharmaceutical compositions that modulate IKZF2
US11897862B2 (en) 2022-03-17 2024-02-13 Gilead Sciences, Inc. IKAROS zinc finger family degraders and uses thereof
WO2023183540A1 (fr) * 2022-03-25 2023-09-28 Regents Of The University Of Michigan Dégradeurs d'ikzf2 et leurs utilisations
WO2023201012A1 (fr) * 2022-04-15 2023-10-19 Regents Of The University Of Michigan Agents de dégradation d'ikzf2 et leurs utilisations
WO2024073475A1 (fr) * 2022-09-27 2024-04-04 Oncopia Therapeutics, Inc. D/B/A Proteovant Therapeutics, Inc. Ligands de cereblon et utilisations associées
CN115252606A (zh) * 2022-09-29 2022-11-01 北京市神经外科研究所 化合物在制备用于***的药物中的应用
CN115252606B (zh) * 2022-09-29 2023-02-03 北京市神经外科研究所 化合物在制备用于***的药物中的应用

Also Published As

Publication number Publication date
AU2020222346B2 (en) 2021-12-09
EP3924055B1 (fr) 2024-04-03
MX2021009764A (es) 2021-09-08
CA3123519A1 (fr) 2020-08-20
US20220144798A1 (en) 2022-05-12
AU2020222346A1 (en) 2021-07-08
EA202192029A1 (ru) 2021-10-27
JP2022520448A (ja) 2022-03-30
KR20210129672A (ko) 2021-10-28
EP3924055A1 (fr) 2021-12-22
CN113329792A (zh) 2021-08-31

Similar Documents

Publication Publication Date Title
AU2019301947B2 (en) 3-(5-amino-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of I KAROS Family Zinc Finger 2 (IKZF2)-dependent diseases
AU2019301944B2 (en) 3-(5-hydroxy-1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and their use in the treatment of IKAROS Family Zinc Finger 2 (IKZF2)-dependent diseases
AU2020222346B2 (en) Substituted 3-(1-oxoisoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
AU2020222345B2 (en) 3-(1-oxo-5-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione derivatives and uses thereof
WO2022029573A1 (fr) Dérivés de 3-(1-oxoisoindolin-2-yl)pipéridine-2,6-dione substitués par hétéroaryle et leurs utilisations
TWI830747B (zh) 3-(5-胺基-1-側氧基異吲哚啉-2-基)哌啶-2,6-二酮衍生物及其用途
BR112021015672A2 (pt) Derivados de 3-(1-oxoisoindolin-2-il)piperidina-2,6-diona substituída e usos dos mesmos
BR112021015783A2 (pt) Derivados de 3-(1-oxo-5-(piperidin-4-il)isoindolin-2-il)piperidina-2,6-diona e usos dos mesmos

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20708627

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3123519

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020222346

Country of ref document: AU

Date of ref document: 20200213

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2021547485

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021015672

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217028320

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020708627

Country of ref document: EP

Effective date: 20210915

ENP Entry into the national phase

Ref document number: 112021015672

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210809